51
|
Tong ZB, Kim H, El Touny L, Simeonov A, Gerhold D. LUHMES Dopaminergic Neurons Are Uniquely Susceptible to Ferroptosis. Neurotox Res 2022; 40:1526-1536. [PMID: 35922689 PMCID: PMC9576307 DOI: 10.1007/s12640-022-00538-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 06/13/2022] [Accepted: 06/27/2022] [Indexed: 02/07/2023]
Abstract
Ferroptosis is a necrotic cell death caused by lipid oxidation that may be responsible for neural degeneration in Parkinson's disease. We assessed whether three neuronal cell lines are sensitive to killing by ferroptosis. Ferroptosis inducer erastin killed LUHMES neurons at sub-micromolar concentrations, whereas neuronal cells derived from SH-SY5Y cells or neural stem cells were at least 50-fold less sensitive. LUHMES differentiated neurons were likewise sensitive to killing by RSL3 or ML210, inhibitors of the glutathione peroxidase 4 enzyme (GPX4) that consumes GSH to detoxify lipid peroxides. Additional assays showed that erastin, RSL3, and ML210 increased lipid peroxide levels, and that LUHMES neurons were protected from both peroxide accumulation and cell death by ferrostatin-1. A possible role of iron was assessed by evaluating the effects of five metal chelators on cytotoxicity of erastin and RSL3. LUHMES neurons were protected from RSL3 by three of the chelators, 2,3-dimercapto-1-propanesulfonic acid (DMPS), deferoxiprone (DFX), and deferiprone (DFP). Collectively, these results demonstrate the vulnerability of LUHMES neurons to ferroptosis by chemical treatments that disrupt glutathione synthesis, lipid peroxide detoxification, or iron metabolism. The same vulnerabilities may occur in CNS neurons, which reportedly generate low levels of GSH and metallothioneins, limiting their ability to neutralize oxidative stresses and toxic metals. These results suggest a rationale and methods to search for environmental toxicants that may exploit these vulnerabilities and promote neurodegenerative diseases.
Collapse
Affiliation(s)
- Zhi-Bin Tong
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Hyunhee Kim
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Lara El Touny
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Anton Simeonov
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - David Gerhold
- Division of Preclinical Innovation, National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA.
| |
Collapse
|
52
|
Naoi M, Maruyama W, Shamoto-Nagai M. Neuroprotective Function of Rasagiline and Selegiline, Inhibitors of Type B Monoamine Oxidase, and Role of Monoamine Oxidases in Synucleinopathies. Int J Mol Sci 2022; 23:ijms231911059. [PMID: 36232361 PMCID: PMC9570229 DOI: 10.3390/ijms231911059] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Revised: 09/09/2022] [Accepted: 09/14/2022] [Indexed: 11/27/2022] Open
Abstract
Synucleinopathies are a group of neurodegenerative disorders caused by the accumulation of toxic species of α-synuclein. The common clinical features are chronic progressive decline of motor, cognitive, behavioral, and autonomic functions. They include Parkinson’s disease, dementia with Lewy body, and multiple system atrophy. Their etiology has not been clarified and multiple pathogenic factors include oxidative stress, mitochondrial dysfunction, impaired protein degradation systems, and neuroinflammation. Current available therapy cannot prevent progressive neurodegeneration and “disease-modifying or neuroprotective” therapy has been proposed. This paper presents the molecular mechanisms of neuroprotection by the inhibitors of type B monoamine oxidase, rasagiline and selegiline. They prevent mitochondrial apoptosis, induce anti-apoptotic Bcl-2 protein family, and pro-survival brain- and glial cell line-derived neurotrophic factors. They also prevent toxic oligomerization and aggregation of α-synuclein. Monoamine oxidase is involved in neurodegeneration and neuroprotection, independently of the catalytic activity. Type A monoamine oxidases mediates rasagiline-activated signaling pathways to induce neuroprotective genes in neuronal cells. Multi-targeting propargylamine derivatives have been developed for therapy in various neurodegenerative diseases. Preclinical studies have presented neuroprotection of rasagiline and selegiline, but beneficial effects have been scarcely presented. Strategy to improve clinical trials is discussed to achieve disease-modification in synucleinopathies.
Collapse
Affiliation(s)
- Makoto Naoi
- Correspondence: ; Tel.: +81-05-6173-1111 (ext. 3494); Fax: +81-561-731-142
| | | | | |
Collapse
|
53
|
Liu X, Wang X, Zhang F, Yao X, Qiao Z, Deng J, Jiao Q, Gong L, Jiang X. Toxic effects of fludioxonil on the growth, photosynthetic activity, oxidative stress, cell morphology, apoptosis, and metabolism of Chlorella vulgaris. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 838:156069. [PMID: 35605851 DOI: 10.1016/j.scitotenv.2022.156069] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 05/09/2022] [Accepted: 05/16/2022] [Indexed: 06/15/2023]
Abstract
Fludioxonil is widely used in the control of crop diseases because of its broad spectrum and high activity, but its presence is now common in waterways proximate to treated areas. This study examined the toxic effects and mechanisms of fludioxonil on the microalgal taxa Chlorella vulgaris. The results showed that fludioxonil limited the growth of C. vulgaris and the median inhibitory concentration at 96 h was 1.87 mg/L. Concentrations of 0.75 and 3 mg/L fludioxonil reduced the content of photosynthetic pigments in algal cells to different degrees. Fludioxonil induced oxidative damage by altering C. vulgaris antioxidant enzyme activities and increasing reactive oxygen species levels. Fludioxonil at 0.75 mg/L significantly increased the activity of antioxidant enzymes. The highest level of activity was 1.60 times that of the control group. Both fludioxonil treatment groups significantly increased ROS levels, with the highest increase being 1.90 times that of the control group. Transmission electron microscope showed that treatment with 3 mg/L fludioxonil for 96 h disrupted cell integrity and changed cell morphology, and flow cytometer analysis showed that fludioxonil induced apoptosis. Changes in endogenous substances indicated that fludioxonil negatively affects C. vulgaris via altered energy metabolism, biosynthesis of amino acids, and unsaturated fatty acids. This study elucidates the effects of fludioxonil on microalgae and the biological mechanisms of its toxicity, providing insights into the importance of the proper management of this fungicide.
Collapse
Affiliation(s)
- Xiang Liu
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Xueting Wang
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Fengwen Zhang
- Tobacco Research Institute of Chinese Academy of Agricultural Sciences (CAAS), Qingdao 266101, PR China
| | - Xiangfeng Yao
- College of Resources and Environment, Shandong Agricultural University, Tai'an 271018, PR China
| | - Zhihua Qiao
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Jiahui Deng
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Qin Jiao
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Luo Gong
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China
| | - Xingyin Jiang
- College of Plant Protection, Shandong Agricultural University, Tai'an 271018, PR China.
| |
Collapse
|
54
|
Kim YM, Choi SY, Hwang O, Lee JY. Pyruvate Prevents Dopaminergic Neurodegeneration and Motor Deficits in the 1-Methyl-4-Phenyl-1,2,3,6-Tetrahydropyridine Model of Parkinson's Disease. Mol Neurobiol 2022; 59:6956-6970. [PMID: 36057709 DOI: 10.1007/s12035-022-03017-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 08/23/2022] [Indexed: 11/26/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by the selective loss of dopamine(DA)rgic neurons in the substantia nigra of the midbrain, and primarily causes motor symptoms. While the pathological cause of PD remains uncertain, oxidative damage, neuroinflammation, and energy metabolic perturbation have been implicated. Pyruvate has been shown neuroprotective in animal models for many neurological disorders, presumably owing to its potent anti-oxidative, anti-inflammatory, and energy metabolic properties. We therefore investigated whether exogenous pyruvate could also protect nigral DA neurons from degeneration and reverse the associated motor deficits in an animal model of PD using the DA neuron-specific toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). MPTP (20 mg/kg) was injected four times every 2 h into the peritoneum of mice, which resulted in a massive loss of DA neurons as well as an increase in neuronal death and cytosolic labile zinc overload. There were rises in inflammatory and oxidative responses, a drop in the striatal DA level, and the emergence of PD-related motor deficits. In comparison, when sodium pyruvate was administered intraperitoneally at a daily dose of 250 mg/kg for 7 days starting 2 h after the final MPTP treatment, significant relief in the MPTP-induced neuropathology, neurodegeneration, DA depletion, and motor symptoms was observed. Equiosmolar dose of NaCl had no neuroprotective effect, and lower doses of sodium pyruvate did not have any statistically significant effects. These findings suggest that pyruvate has therapeutic potential for the treatment of PD and related neurodegenerative diseases.
Collapse
Affiliation(s)
- Yun-Mi Kim
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Su Yeon Choi
- Department of Medical Science, Asan Medical Institute of Convergence Science and Technology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea
| | - Onyou Hwang
- Department of Biochemistry and Molecular Biology, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| | - Joo-Yong Lee
- Asan Institute for Life Sciences, Asan Medical Center, Seoul, 05505, Republic of Korea.
- Department of Convergence Medicine, University of Ulsan College of Medicine, Seoul, 05505, Republic of Korea.
| |
Collapse
|
55
|
Keeney MT, Hoffman EK, Farmer K, Bodle CR, Fazzari M, Zharikov A, Castro SL, Hu X, Mortimer A, Kofler JK, Cifuentes-Pagano E, Pagano PJ, Burton EA, Hastings TG, Greenamyre JT, Di Maio R. NADPH oxidase 2 activity in Parkinson's disease. Neurobiol Dis 2022; 170:105754. [PMID: 35577065 PMCID: PMC9284948 DOI: 10.1016/j.nbd.2022.105754] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/09/2022] [Accepted: 05/10/2022] [Indexed: 11/24/2022] Open
Abstract
Mitochondrial dysfunction and oxidative stress are strongly implicated in Parkinson's disease (PD) pathogenesis and there is evidence that mitochondrially-generated superoxide can activate NADPH oxidase 2 (NOX2). Although NOX2 has been examined in the context of PD, most attention has focused on glial NOX2, and the role of neuronal NOX2 in PD remains to be defined. Additionally, pharmacological NOX2 inhibitors have typically lacked specificity. Here we devised and validated a proximity ligation assay for NOX2 activity and demonstrated that in human PD and two animal models thereof, both neuronal and microglial NOX2 are highly active in substantia nigra under chronic conditions. However, in acute and sub-acute PD models, we observed neuronal, but not microglial NOX2 activation, suggesting that neuronal NOX2 may play a primary role in the early stages of the disease. Aberrant NOX2 activity is responsible for the formation of oxidative stress-related post-translational modifications of α-synuclein, and impaired mitochondrial protein import in vitro in primary ventral midbrain neuronal cultures and in vivo in nigrostriatal neurons in rats. In a rat model, administration of a brain-penetrant, highly specific NOX2 inhibitor prevented NOX2 activation in nigrostriatal neurons and its downstream effects in vivo, such as activation of leucine-rich repeat kinase 2 (LRRK2). We conclude that NOX2 is an important enzyme that contributes to progressive oxidative damage which in turn can lead to α-synuclein accumulation, mitochondrial protein import impairment, and LRRK2 activation. In this context, NOX2 inhibitors hold potential as a disease-modifying therapy in PD.
Collapse
Affiliation(s)
- Matthew T Keeney
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eric K Hoffman
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kyle Farmer
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Christopher R Bodle
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Marco Fazzari
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Alevtina Zharikov
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Sandra L Castro
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoping Hu
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Amanda Mortimer
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Julia K Kofler
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Eugenia Cifuentes-Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick J Pagano
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Vascular Medicine Institute, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Edward A Burton
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; Geriatric Research, Education and Clinical Center, VA Pittsburgh Healthcare System, Pittsburgh, PA 15240, USA
| | - Teresa G Hastings
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - J Timothy Greenamyre
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Roberto Di Maio
- Pittsburgh Institute for Neurodegenerative Diseases, Pittsburgh, PA 15213, USA; Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA.
| |
Collapse
|
56
|
Naked mole-rats resist the accumulation of hypoxia-induced oxidative damage. Comp Biochem Physiol A Mol Integr Physiol 2022; 273:111282. [PMID: 35907588 DOI: 10.1016/j.cbpa.2022.111282] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 01/28/2023]
Abstract
Naked mole-rats are among the few mammals with the ability to endure severe hypoxia. These unique rodents use metabolic rate depression along with various molecular mechanisms to successfully overcome the challenges of oxygen-limitation, which they experience in their underground borrows. While studies have reported that naked mole-rats exhibit inherently higher levels of oxidative damage across their lifespan as compared to mice, it has yet to be determined whether naked mole-rats are vulnerable to oxidative damage during periods of low oxygen exposure. To investigate this phenomenon, we examined cellular oxidative damage markers of macromolecules: DNA oxidation determined as 8-oxo-2'deoxyguanosine (8-OHdG8) levels, RNA oxidation as 8-hydroxyguanosine (8-OHG), protein carbonylation, and lipid peroxidation in normoxic (control), acute (4 h at 7% O2), and chronic (24 h at 7% O2) hypoxia-exposed naked mole-rats. Brain appears to be the most resilient organ to hypoxia-induced oxidative damage, with both brain and heart exhibiting enhanced antioxidant capacity during hypoxia. Levels of DNA and RNA oxidation were minimally changed in all tissues and no changes were observed in protein carbonylation. Most tissues experienced lipid peroxidation, with liver displaying a 9.6-fold increase during hypoxia. Concomitantly, levels of DNA damage repair proteins were dynamically regulated in a tissue-specific manner, with white adipose displaying a significant reduction during hypoxia. Our findings show that naked mole-rats largely avoid hypoxia-induced oxidative damage, possibly due to their high tolerance to redox stress, or to reduced oxidative requirements made possible during their hypometabolic response when oxygen supply is limited.
Collapse
|
57
|
Liu MM, Zhou N, Jiang N, Lu KM, Wu CF, Bao JK. Neuroprotective Effects of Oligosaccharides From Periplaneta Americana on Parkinson’s Disease Models In Vitro and In Vivo. Front Pharmacol 2022; 13:936818. [PMID: 35924055 PMCID: PMC9340460 DOI: 10.3389/fphar.2022.936818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Accepted: 05/31/2022] [Indexed: 11/22/2022] Open
Abstract
Parkinson’s disease (PD) is one of the neurodegenerative diseases that is characterized by obvious motor and some nonmotor symptoms. Various therapeutics failed in the effective treatment of PD because of impaired neurological function in the brain and various complications. Periplaneta Americana oligosaccharides (OPA), the main active ingredients extracted from the medicine residues of Periplaneta Americana (P. Americana), have been reported to exert anti-inflammatory effects. The purpose of this study was to evaluate the possible mechanisms of OPA against 1-methyl-4-phenylpyridinium (MPP+)-induced apotosis in SH-SY5Y cells and its potential neuroprotective effects in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced PD subacute model mice. The data demonstrated that OPA significantly reversed the MPP+-induced decrease in SH-SY5Y cell viability, reduced the proportion of apoptotic cells, and protected SH-SY5Y cells from apoptosis in a dose-dependent manner by regulating the expression of apoptosis-related genes. Furthermore, OPA also alleviated the motor dysfunction of PD model mice, prevented the loss of tyrosine hydroxylase positive cells, suppressed the apoptosis of substantia nigra cells, and improved the dysbiosis of gut microbiota in vivo, suggesting that OPA demonstrated a significantly neuroprotective effect on PD model mice. These results indicated that OPA might be the possibility of PD therapeutics with economic utility and high safety.
Collapse
Affiliation(s)
- Miao-Miao Liu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Nan Zhou
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Na Jiang
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
| | - Kai-Min Lu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- Pharmacy Research Center, Binzhou Medical University, Yantai, China
| | - Chuan-Fang Wu
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Chuan-Fang Wu, ; Jin-Ku Bao,
| | - Jin-Ku Bao
- Key Laboratory of Bio-Resource and Eco-Environment of Ministry of Education, College of Life Sciences, Sichuan University, Chengdu, China
- *Correspondence: Chuan-Fang Wu, ; Jin-Ku Bao,
| |
Collapse
|
58
|
Singh S, Ganguly U, Pal S, Chandan G, Thakur R, Saini RV, Chakrabarti SS, Agrawal BK, Chakrabarti S. Protective effects of cyclosporine A on neurodegeneration and motor impairment in rotenone-induced experimental models of Parkinson's disease. Eur J Pharmacol 2022; 929:175129. [PMID: 35777442 DOI: 10.1016/j.ejphar.2022.175129] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 11/30/2022]
Abstract
The development of neuroprotective drugs targeting mitochondria could be an important strategy in combating the progressive clinical course of Parkinson's disease. In the current study, we demonstrated that in SH-SY5Y cells (human dopaminergic neuroblastoma cell line), rotenone caused a dose-dependent (0.25-1 μM) and time-dependent (up to 48 h) loss of cell viability and a loss of cellular ATP content with mitochondrial membrane depolarization and an increased formation of reactive oxygen species; all these processes were markedly prevented by the mitochondrial permeability transition pore blocker cyclosporine A, which did not affect complex I inhibition by rotenone. The nuclear morphology of rotenone-treated cells for 48 h indicated the presence of both necrosis and apoptosis. We then examined the effects of cyclosporine A on the rotenone-induced model of Parkinson's disease in Wistar rats. Cyclosporine A significantly improved the motor deficits and prevented the loss of nigral dopaminergic neurons projecting into the striatum in rotenone-treated rats. Being a marketed immuno-suppressive drug, cyclosporine A should be further evaluated for its putative neuroprotective action in Parkinson's disease.
Collapse
Affiliation(s)
- Sukhpal Singh
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Upasana Ganguly
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Soumya Pal
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India; Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Gourav Chandan
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Rahul Thakur
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Reena V Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Sankha Shubhra Chakrabarti
- Department of Geriatric Medicine, Institute of Medical Sciences, Banaras Hindu University, Varanasi, India
| | - Bimal K Agrawal
- Department of Medicine, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Cell, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar University (Deemed to be), Mullana, Ambala, India.
| |
Collapse
|
59
|
Dhillon T, Kumar A, Kumar V. Neuroprotective Effect of N-acetylcysteine Against Monocrotophos-Induced Oxidative Stress in Different Brain Regions of Rats. Appl Biochem Biotechnol 2022; 194:4049-4065. [PMID: 35587328 DOI: 10.1007/s12010-022-03967-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Accepted: 05/10/2022] [Indexed: 11/02/2022]
Abstract
Monocrotophos (MCP) is systemic organophosphate insecticide used against crop pests. It is reported to cause mammalian toxicity through both acute and chronic exposure. In the present study, we have shown the protective role of N-acetylcysteine (NAC) against MCP-induced oxidative stress in frontal cortex, corpus striatum and hippocampus brain regions of rats. Male Albino Wistar rats were divided into control, NAC-treated, MCP and NAC + MCP-treated groups. An oral dose of MCP (0.9 mg/kg b.wt) and NAC (200 mg/kg b.wt) was administered for 28 days. Results showed an increase in lipid peroxidation (LPO) and protein oxidation followed by decreased antioxidant enzymes after 28 days of MCP exposure. Histopathological analysis showed that monocrotophos exposure caused neurodegenerative changes as evident by neurons with dystrophic changes in the form of shrunken hyperchromatic nuclei in all the regions of the rat brain. N-acetylcysteine supplementation to MCP-treated rats showed a reduction in oxidative stress and ameliorated cellular alterations in all of the three regions. The results of the study indicate that N-acetylcysteine offers neuroprotection by improving antioxidant response and decreasing oxidative stress in different regions of the rat brain.
Collapse
Affiliation(s)
- Twinkle Dhillon
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Amit Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India
| | - Vijay Kumar
- Department of Biochemistry, Maharshi Dayanand University, Rohtak, 124001, Haryana, India.
| |
Collapse
|
60
|
Ayed-Boussema I, Hamdi H, Chaabani H, M’nassri A, Mokni M, Abid S. Fenpyroximate induced cytotoxicity and genotoxicity in Wistar rat brain and in human neuroblastoma (SH-SY5Y) cells: involvement of oxidative stress and apoptosis. Neurotoxicology 2022; 91:177-187. [DOI: 10.1016/j.neuro.2022.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 04/21/2022] [Accepted: 05/11/2022] [Indexed: 11/16/2022]
|
61
|
Zavadskiy S, Sologova S, Moldogazieva N. Oxidative distress in aging and age-related diseases: Spatiotemporal dysregulation of protein oxidation and degradation. Biochimie 2022; 195:114-134. [PMID: 34890732 DOI: 10.1016/j.biochi.2021.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 12/03/2021] [Accepted: 12/04/2021] [Indexed: 12/31/2022]
Abstract
The concept of oxidative distress had arisen from the assessment of cellular response to high concentrations of reactive species that result from an imbalance between oxidants and antioxidants and cause biomolecular damage. The intracellular distribution and flux of reactive species dramatically change in time and space contributing to the remodeling of the redox landscape and sensitivity of protein residues to oxidants. Here, we hypothesize that compromised spatiotemporal control of generation, conversions, and removal of reactive species underlies protein damage and dysfunction of protein degradation machineries. This leads to the accumulation of oxidatively damaged proteins resulted in an age-dependent decline in the organismal adaptability to oxidative stress. We highlight recent data obtained with the use of various cell cultures, animal models, and patients on irreversible and non-repairable oxidation of key redox-sensitive residues. Multiple reaction products include peptidyl hydroperoxides, alcohols, carbonyls, and carbamoyl moieties as well as Tyr-Tyr, Trp-Tyr, Trp-Trp, Tyr-Cys, His-Lys, His-Arg, and Tyr-Lys cross-links. These lead to protein fragmentation, misfolding, covalent cross-linking, oligomerization, aggregation, and ultimately, causing impaired protein function and turnover. 20S proteasome and autophagy-lysosome pathways are two major types of machinery for the degradation and elimination of oxidatively damaged proteins. Spatiotemporal dysregulation of these pathways under oxidative distress conditions is implicated in aging and age-related disorders such as neurodegenerative and cardiovascular diseases and diabetes. Future investigations in this field allow the discovery of new drugs to target components of dysregulated cell signaling and protein degradation machinery to combat aging and age-related chronic diseases.
Collapse
Affiliation(s)
- Sergey Zavadskiy
- Department of Pharmacology, Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Sechenov University, 119991, Moscow, Russia
| | - Susanna Sologova
- Department of Pharmacology, Nelyubin Institute of Pharmacy, I.M. Sechenov First Moscow State Medical University, Sechenov University, 119991, Moscow, Russia
| | - Nurbubu Moldogazieva
- Laboratory of Bioinformatics, Institute of Translational Medicine and Biotechnology, I.M. Sechenov First Moscow State Medical University, Sechenov University, 119991, Moscow, Russia.
| |
Collapse
|
62
|
Neonatal 6-hydroxydopamine lesioning of rats and dopaminergic neurotoxicity: proposed animal model of Parkinson’s disease. J Neural Transm (Vienna) 2022; 129:445-461. [DOI: 10.1007/s00702-022-02479-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 02/11/2022] [Indexed: 10/18/2022]
|
63
|
Onaolapo OJ, Odeniyi AO, Onaolapo AY. Parkinson's Disease: Is there a Role for Dietary and Herbal Supplements? CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2022; 20:343-365. [PMID: 33602107 DOI: 10.2174/1871527320666210218082954] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 09/19/2020] [Accepted: 09/29/2020] [Indexed: 12/18/2022]
Abstract
Parkinson's Disease (PD) is characterised by degeneration of the neurons of the nigrostriatal dopaminergic pathway of the brain. The pharmacological cornerstone of PD management is mainly the use of dopamine precursors, dopamine receptor agonists, and agents that inhibit the biochemical degradation of dopamine. While these drugs initially provide relief to the symptoms and improve the quality of life of the patients, progression of the underlying pathological processes, such as oxidative stress and neuroinflammation (which have been strongly associated with PD and other neurodegenerative disorders), eventually reduce their benefits, making further benefits achievable, only at high doses due to which the magnitude and frequency of side-effects are amplified. Also, while it is becoming obvious that mainstream pharmacological agents may not always provide the much-needed answer, the question remains what succour can nature provide through dietary supplements, nutraceuticals and herbal remedies? This narrative review examines current literature for evidence of the possible roles (if any) of nutraceuticals, dietary supplements and herbal remedies in the prevention or management of PD by examining how these compounds could modulate key factors and pathways that are crucial to the pathogenesis and/or progression of PD. The likely limitations of this approach and its possible future roles in PD prevention and management are also considered.
Collapse
Affiliation(s)
- Olakunle J Onaolapo
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Ademola O Odeniyi
- Behavioural Neuroscience Unit, Neuropharmacology Subdivision, Department of Pharmacology, Ladoke Akintola University of Technology, Osogbo, Osun State, Nigeria
| | - Adejoke Y Onaolapo
- Behavioural Neuroscience Unit, Neurobiology Subdivision, Department of Anatomy, Ladoke Akintola University of Technology, Ogbomosho, Oyo State, Nigeria
| |
Collapse
|
64
|
Couto SDF, Araujo SM, Bortolotto VC, Dahleh MMM, Musachio EAS, Pinheiro FC, Romio LC, do Sacramento M, Alves D, Prigol M. Effectiveness of 7-chloro-4-(phenylselanyl) quinoline in improving learning, short-term memory, and anxiety-like behaviors in a mimetic model of Parkinson's disease in Drosophila melanogaster. NEW J CHEM 2022. [DOI: 10.1039/d2nj04011e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
The potential of 4-PSQ on psychomotor and non-motor behaviors of PD, such as spontaneous locomotor activity, learning, memory, and anxiety.
Collapse
Affiliation(s)
- Shanda de Freitas Couto
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
- Departamento de Nutrição – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Stífani Machado Araujo
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Mustafa Munir Mustafa Dahleh
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Franciane Cabral Pinheiro
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Leugim Corteze Romio
- Departamento de Matemática – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| | - Manoela do Sacramento
- Laboratório de Síntese Orgânica Limpa – LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA) – Universidade Federal de Pelotas – Campus Universitário, S/N – Prédio/Bloco: 30 e 32, Capão do Leão, Rio Grande do Sul, CEP 96160-000, Brazil
| | - Diego Alves
- Laboratório de Síntese Orgânica Limpa – LASOL, Centro de Ciências Químicas, Farmacêuticas e de Alimentos (CCQFA) – Universidade Federal de Pelotas – Campus Universitário, S/N – Prédio/Bloco: 30 e 32, Capão do Leão, Rio Grande do Sul, CEP 96160-000, Brazil
| | - Marina Prigol
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas – LaftamBio Pampa – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
- Departamento de Nutrição – Universidade Federal do Pampa – Campus Itaqui – Rua Luiz Joaquim de Sá Britto, s/n – Bairro: Promorar, Itaqui, Rio Grande do Sul, CEP 97650-000, Brazil
| |
Collapse
|
65
|
Vitamin B12 Reduces TDP-43 Toxicity by Alleviating Oxidative Stress and Mitochondrial Dysfunction. Antioxidants (Basel) 2021; 11:antiox11010082. [PMID: 35052586 PMCID: PMC8773243 DOI: 10.3390/antiox11010082] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/23/2021] [Accepted: 12/26/2021] [Indexed: 01/20/2023] Open
Abstract
TAR DNA-binding protein 43 (TDP-43) is a member of an evolutionarily conserved family of heterogeneous nuclear ribonucleoproteins that modulate multiple steps in RNA metabolic processes. Cytoplasmic aggregation of TDP-43 in affected neurons is a pathological hallmark of many neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), frontotemporal dementia (FTD), Alzheimer’s disease (AD), and limbic predominant age-related TDP-43 encephalopathy (LATE). Mislocalized and accumulated TDP-43 in the cytoplasm induces mitochondrial dysfunction and reactive oxidative species (ROS) production. Here, we show that TDP-43- and rotenone-induced neurotoxicity in the human neuronal cell line SH-SY5Y were attenuated by hydroxocobalamin (Hb, vitamin B12 analog) treatment. Although Hb did not affect the cytoplasmic accumulation of TDP-43, Hb attenuated TDP-43-induced toxicity by reducing oxidative stress and mitochondrial dysfunction. Moreover, a shortened lifespan and motility defects in TDP-43-expressing Drosophila were significantly mitigated by dietary treatment with hydroxocobalamin. Taken together, these findings suggest that oral intake of hydroxocobalamin may be a potential therapeutic intervention for TDP-43-associated proteinopathies.
Collapse
|
66
|
Moulton MJ, Barish S, Ralhan I, Chang J, Goodman LD, Harland JG, Marcogliese PC, Johansson JO, Ioannou MS, Bellen HJ. Neuronal ROS-induced glial lipid droplet formation is altered by loss of Alzheimer's disease-associated genes. Proc Natl Acad Sci U S A 2021; 118:e2112095118. [PMID: 34949639 PMCID: PMC8719885 DOI: 10.1073/pnas.2112095118] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/11/2021] [Indexed: 01/02/2023] Open
Abstract
A growing list of Alzheimer's disease (AD) genetic risk factors is being identified, but the contribution of each variant to disease mechanism remains largely unknown. We have previously shown that elevated levels of reactive oxygen species (ROS) induces lipid synthesis in neurons leading to the sequestration of peroxidated lipids in glial lipid droplets (LD), delaying neurotoxicity. This neuron-to-glia lipid transport is APOD/E-dependent. To identify proteins that modulate these neuroprotective effects, we tested the role of AD risk genes in ROS-induced LD formation and demonstrate that several genes impact neuroprotective LD formation, including homologs of human ABCA1, ABCA7, VLDLR, VPS26, VPS35, AP2A, PICALM, and CD2AP Our data also show that ROS enhances Aβ42 phenotypes in flies and mice. Finally, a peptide agonist of ABCA1 restores glial LD formation in a humanized APOE4 fly model, highlighting a potentially therapeutic avenue to prevent ROS-induced neurotoxicity. This study places many AD genetic risk factors in a ROS-induced neuron-to-glia lipid transfer pathway with a critical role in protecting against neurotoxicity.
Collapse
Affiliation(s)
- Matthew J Moulton
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Scott Barish
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
| | - Isha Ralhan
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Jinlan Chang
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Lindsey D Goodman
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Jake G Harland
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | - Paul C Marcogliese
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
| | | | - Maria S Ioannou
- Department of Physiology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Group on Molecular and Cell Biology of Lipids, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Department of Cell Biology, University of Alberta, Edmonton, AB T6G 2R3, Canada
- Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB T6G 2R3, Canada
| | - Hugo J Bellen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030;
- Jan and Dan Duncan Neurological Research Institute, Texas Children's Hospital, Houston, TX 77030
- Program in Developmental Biology, Baylor College of Medicine, Houston TX 77030
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030
| |
Collapse
|
67
|
Milosavljevic S, Djordjevic I, Mandic B, Tesevic V, Stankovic M, Todorovic N, Novakovic M. Flavonoids of the Heartwood of Cotinus coggygria Scop. Showing Protective Effect on Human Lymphocyte DNA. Nat Prod Commun 2021. [DOI: 10.1177/1934578x211067289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
In continuation of our study on Cotinus coggygria from Serbia, 10 known flavonoids (1-10) were isolated from the methylene chloride/methanol extract of the heartwood. They were tested for in vitro protective effect against chromosome aberrations in peripheral human lymphocytes, using the cytokinesis-block micronucleus assay. All tested compounds (in minimal doses of 1 μg/mL) exerted a beneficial effect by decreasing DNA damage of human lymphocytes in the range of 24.2% to 54.5%, better than the radio protectant control, amifostine. Functional groups, such as 3′,4′-dihydroxyphenyl (catechol), 5-OH, 3-OH, and 4-keto in flavonoids (3-keto in aurones), which play a key role in antioxidant activity, are proposed to be responsible for the DNA protective activity of the tested compounds.
Collapse
Affiliation(s)
| | - Iris Djordjevic
- Faculty of Veterinary Medicine, University of Belgrade, Belgrade, Serbia
| | - Boris Mandic
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Vele Tesevic
- Faculty of Chemistry, University of Belgrade, Belgrade, Serbia
| | | | - Nina Todorovic
- Instituite of Chemistry, Technology and Metallurgy, Department of Chemistry, University of Belgrade, Belgrade, Serbia
| | - Miroslav Novakovic
- Faculty of Veterinary Medicine, University of Belgrade, Belgrade, Serbia
- Dedicated to Professor Yoshinori Asakawa on the occasion of his 80th birthday
| |
Collapse
|
68
|
Anesthesia-Induced Oxidative Stress: Are There Differences between Intravenous and Inhaled Anesthetics? OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8782387. [PMID: 34873432 PMCID: PMC8643269 DOI: 10.1155/2021/8782387] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/11/2021] [Accepted: 11/12/2021] [Indexed: 12/11/2022]
Abstract
Agents used for the induction of anesthesia have been shown to either promote or mitigate oxidative stress. A fine balance between the presence of reactive oxygen species (ROS) and antioxidants is crucial for the proper normal functioning of the cell. A basal concentration of ROS is essential for the manifestation of cellular functions, whereas disproportionate levels of ROS cause damage to cellular macromolecules such as DNA, lipids, and proteins, eventually leading to necrosis and apoptosis. Increased ROS has been linked with numerous illnesses, such as cardiovascular, immune system, liver, and kidney, and has been shown to promote cancer and accelerate aging. Knowledge of the various pharmacologic agents that increase or reduce oxidative stress may promote a safer way of inducing anesthesia. Furthermore, surgery itself leads to increased ROS production and ischemia/reperfusion injury. Indeed, increased perioperative oxidative stress has been correlated with increased postoperative complications and prolonged recovery. Anesthesiologists care for patients during the whole spectrum of perioperative care and thus are in a unique position to deliver countermeasures to oxidative stress. Using preferentially an induction agent which reduces oxidative stress might lead to better clinical outcomes and fewer postoperative complications. Propofol has been shown in several studies to reduce oxidative stress, which reduces postoperative complications and leads to a faster recovery, and thus might represent the preferred induction agent in the right clinical setting.
Collapse
|
69
|
Li C, Zhang Y, Liu R, Mai Y. Ramelteon ameliorated 1-methyl-4-phenylpyridinium (MPP+)-induced neurotoxicity in neuronal cells in a mitochondrial-dependent pathway. Bioengineered 2021; 12:4868-4877. [PMID: 34346829 PMCID: PMC8806746 DOI: 10.1080/21655979.2021.1960767] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 07/22/2021] [Indexed: 12/25/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease with global health and economic impact. 1-methyl-4-phenylpyridinium (MPP+)-induced mitochondrial dysfunction and oxidative stress are reported to participate in the pathological mechanism of PD. Ramelteon is a novel oral hypnotic agent that has recently been reported to display neuronal protective effects. However, it is unknown whether Ramelteon possesses a beneficial effect in PD. In this study, we aimed to examine the potential function of Ramelteon in MPP+-challenged neurons. We found that Ramelteon rescued the cell viability reduced by MPP+-stimulation. Further, oxidative stress in MPP+-challenged SH-SY5Y cells was mitigated by Ramelteon as verified by the upregulated levels of mitochondrial reactive oxygen species (ROS) and protein carboxyl, and the upregulation of NADPH oxidase 4 (NOX-4). Furthermore, the declined mitochondrial membrane potential (ΔΨm) caused by MPP+ was reversed by Ramelteon. Importantly, Ramelteon attenuated MPP+-induced apoptosis, accompanied by a decreased ratio of Bax/Bcl-2, inhibition of cytochrome C release, and downregulation of cleaved caspase-3. For the first time, we conclude that Ramelteon might ameliorate MPP+-induced neurotoxicity in neuronal cells in a mitochondrial-dependent pathway.
Collapse
Affiliation(s)
- Chuo Li
- Department of Neurology, Guangzhou Eighth People’s Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yusheng Zhang
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Rongrong Liu
- Department of Neurology and Stroke Center, The First Affiliated Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yuzhen Mai
- Department of Neurology, Guangzhou Eighth People’s Hospital of Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
70
|
de Rus Jacquet A, Ambaw A, Tambe MA, Ma SY, Timmers M, Grace MH, Wu QL, Simon JE, McCabe GP, Lila MA, Shi R, Rochet JC. Neuroprotective mechanisms of red clover and soy isoflavones in Parkinson's disease models. Food Funct 2021; 12:11987-12007. [PMID: 34751296 PMCID: PMC10822195 DOI: 10.1039/d1fo00007a] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by nigrostriatal degeneration and the spreading of aggregated forms of the presynaptic protein α-synuclein (aSyn) throughout the brain. PD patients are currently only treated with symptomatic therapies, and strategies to slow or stop the progressive neurodegeneration underlying the disease's motor and cognitive symptoms are greatly needed. The time between the first neurobiochemical alterations and the initial presentation of symptoms is thought to span several years, and early neuroprotective dietary interventions could delay the disease onset or slow PD progression. In this study, we characterized the neuroprotective effects of isoflavones, a class of dietary polyphenols found in soy products and in the medicinal plant red clover (Trifolium pratense). We found that isoflavone-rich extracts and individual isoflavones rescued the loss of dopaminergic neurons and the shortening of neurites in primary mesencephalic cultures exposed to two PD-related insults, the environmental toxin rotenone and an adenovirus encoding the A53T aSyn mutant. The extracts and individual isoflavones also activated the Nrf2-mediated antioxidant response in astrocytes via a mechanism involving inhibition of the ubiquitin-proteasome system, and they alleviated deficits in mitochondrial respiration. Furthermore, an isoflavone-enriched soy extract reduced motor dysfunction exhibited by rats lesioned with the PD-related neurotoxin 6-OHDA. These findings suggest that plant-derived isoflavones could serve as dietary supplements to delay PD onset in at-risk individuals and mitigate neurodegeneration in the brains of patients.
Collapse
Affiliation(s)
- Aurélie de Rus Jacquet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
| | - Abeje Ambaw
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
| | - Mitali Arun Tambe
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
| | - Sin Ying Ma
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
| | - Michael Timmers
- Plants for Human Health Institute, Department of Food Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, 28081, USA
| | - Mary H Grace
- Plants for Human Health Institute, Department of Food Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, 28081, USA
| | - Qing-Li Wu
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, 08901, USA
| | - James E Simon
- Department of Plant Biology, Rutgers University, New Brunswick, NJ, 08901, USA
| | - George P McCabe
- Department of Statistics, Purdue University, West Lafayette, IN 47907, USA
| | - Mary Ann Lila
- Plants for Human Health Institute, Department of Food Bioprocessing and Nutrition Sciences, North Carolina State University, Kannapolis, NC, 28081, USA
| | - Riyi Shi
- Department of Basic Medical Sciences, Purdue University, West Lafayette, IN, 47907, USA
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, 47907, USA
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Jean-Christophe Rochet
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, 47907, USA.
- Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| |
Collapse
|
71
|
Vahdati SN, Lashkari A, Navasatli SA, Ardestani SK, Safavi M. Butylated hydroxyl-toluene, 2,4-Di-tert-butylphenol, and phytol of Chlorella sp. protect the PC12 cell line against H 2O 2-induced neurotoxicity. Biomed Pharmacother 2021; 145:112415. [PMID: 34775236 DOI: 10.1016/j.biopha.2021.112415] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2021] [Revised: 10/21/2021] [Accepted: 11/05/2021] [Indexed: 12/12/2022] Open
Abstract
Oxidative stress is considered the main cause of cellular damage in a number of neurodegenerative disorders. One suitable ways to prevent cell damage is the use of the exogenous antioxidant capacity of natural products, such as microalgae. In the present study, four microalgae extracts, isolated from the Persian Gulf, were screened to analyze their potential antioxidant activity and free radical scavenging using ABTS, DPPH, and FRAP methods. The methanolic extracts (D1M) of green microalgae derived from Chlorella sp. exhibited potent free radical scavenging activity. In order to characterize microalgae species, microscopic observations and analysis of the expression of 18S rRNA were performed. The antioxidant and neuroprotective effects of D1M on H2O2-induced toxicity in PC12 cells were investigated. The results demonstrated that D1M significantly decreased the release of nitric oxide (NO), formation of intracellular reactive oxygen species (ROS), and the level of malondialdehyde (MDA), whereas it enhanced the content of glutathione (GSH), and activity of heme oxygenase 1 (HO-1), NAD(P)H: quinone oxidoreductase 1 (NQO1), and catalase (CAT) in PC12 cells exposed to H2O2. The pretreatment of D1M improved cell viability as measured by the MTT assay and invert microscopy, reduced cell apoptosis as examined by flow cytometry analysis, increased mitochondrial membrane potential (MMP), and diminished caspase-3 activity. The GC/MS analysis revealed that D1M ingredients have powerful antioxidant and anti-inflammatory compounds, such as butylated hydroxytoluene (BHT), 2,4-di-tert-butyl-phenol (2,4-DTBP), and phytol. These results suggested that Chlorella sp. extracts have strong potential to be applied as neuroprotective agents, for the treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Saeed Niazi Vahdati
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Ali Lashkari
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Sepideh Aliniaye Navasatli
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Susan Kabudanian Ardestani
- Institute of Biochemistry and Biophysics, Department of Biochemistry, University of Tehran, Tehran, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran, Iran.
| |
Collapse
|
72
|
Simões RF, Pino R, Moreira-Soares M, Kovarova J, Neuzil J, Travasso R, Oliveira PJ, Cunha-Oliveira T, Pereira FB. Quantitative analysis of neuronal mitochondrial movement reveals patterns resulting from neurotoxicity of rotenone and 6-hydroxydopamine. FASEB J 2021; 35:e22024. [PMID: 34751984 DOI: 10.1096/fj.202100899r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 09/24/2021] [Accepted: 10/19/2021] [Indexed: 01/31/2023]
Abstract
Alterations in mitochondrial dynamics, including their intracellular trafficking, are common early manifestations of neuronal degeneration. However, current methodologies used to study mitochondrial trafficking events rely on parameters that are primarily altered in later stages of neurodegeneration. Our objective was to establish a reliable applied statistical analysis to detect early alterations in neuronal mitochondrial trafficking. We propose a novel quantitative analysis of mitochondria trajectories based on innovative movement descriptors, including straightness, efficiency, anisotropy, and kurtosis. We evaluated time- and dose-dependent alterations in trajectory descriptors using biological data from differentiated SH-SY5Y cells treated with the mitochondrial toxicants 6-hydroxydopamine and rotenone. MitoTracker Red CMXRos-labelled mitochondria movement was analyzed by total internal reflection fluorescence microscopy followed by computational modelling to describe the process. Based on the aforementioned trajectory descriptors, this innovative analysis of mitochondria trajectories provides insights into mitochondrial movement characteristics and can be a consistent and sensitive method to detect alterations in mitochondrial trafficking occurring in the earliest time points of neurodegeneration.
Collapse
Affiliation(s)
- Rui F Simões
- CNC, Center for Neuroscience and Cell Biology, UC Biotech, Cantanhede, Portugal
| | - Rute Pino
- CISUC, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal
| | - Maurício Moreira-Soares
- OCBE, Faculty of Medicine, University of Oslo, Oslo, Norway.,Centre for Bioinformatics, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo, Norway
| | - Jaromira Kovarova
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, Prague-West, Czech Republic.,School of Medical Science, Griffith University, Southport, Queensland, Australia
| | - Rui Travasso
- CFisUC, Department of Physics, University of Coimbra, Coimbra, Portugal
| | - Paulo J Oliveira
- CNC, Center for Neuroscience and Cell Biology, UC Biotech, Cantanhede, Portugal
| | | | - Francisco B Pereira
- CISUC, Department of Informatics Engineering, University of Coimbra, Coimbra, Portugal.,Coimbra Polytechnic - ISEC, Coimbra, Portugal
| |
Collapse
|
73
|
Queiroz Junior NF, Steffani JA, Machado L, Longhi PJH, Montano MAE, Martins M, Machado SA, Machado AK, Cadoná FC. Antioxidant and cytoprotective effects of avocado oil and extract ( Persea americana Mill) against rotenone using monkey kidney epithelial cells (Vero). JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2021; 84:875-890. [PMID: 34256683 DOI: 10.1080/15287394.2021.1945515] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Oxidative stress is known to be involved in development of numerous diseases including cardiovascular, respiratory, renal, kidney and cancer. Thus, investigations that mimic oxidative stress in vitro may play an important role to find new strategies to control oxidative stress and subsequent consequences are important. Rotenone, widely used as a pesticide has been used as a model to simulate oxidative stress. However, this chemical was found to produce several diseases. Therefore, the aim of this study was to investigate the antioxidant and cytoprotective effect of avocado (Persea americana Mill) extract and oil in monkey kidney epithelial cells (VERO) exposed to rotenone. VERO cells were exposed to IC50 of rotenone in conjunction with different concentrations of avocado extract and oil (ranging from 1 to 1000 µg/ml), for 24 hr. Subsequently, cell viability and oxidative metabolism were assessed. Data demonstrated that avocado extract and oil in the presence of rotenone increased cellular viability at all tested concentrations compared to cells exposed only to rotenone. In addition, extract and avocado oil exhibited antioxidant action as evidenced by decreased levels of reactive oxygen species (ROS), superoxide ion, and lipid peroxidation, generated by rotenone. Further, avocado extract and oil appeared to be safe, since these compounds did not affect cell viability and or generate oxidative stress. Therefore, avocado appears to display a promising antioxidant potential by decreasing oxidative stress.
Collapse
Affiliation(s)
| | - Jovani Antônio Steffani
- Postgraduate Program of Biosciences and Health, West University of Santa Catarina, Joaçaba, SC, Brazil
| | - Larissa Machado
- Biological Sciences Course, West University of Santa Catarina, Joaçaba, SC, Brazil
| | | | | | - Mathias Martins
- Postgraduate Program in Health and Animal Production, West University of Santa Catarina, Joaçaba, SC, Brazil
| | - Sérgio Abreu Machado
- Postgraduate Program in Health and Animal Production, West University of Santa Catarina, Joaçaba, SC, Brazil
| | | | - Francine Carla Cadoná
- Postgraduate Program in Sciences of Health and Life, Franciscan University, Santa Maria, RS, Brazil
| |
Collapse
|
74
|
Mani S, Sevanan M, Krishnamoorthy A, Sekar S. A systematic review of molecular approaches that link mitochondrial dysfunction and neuroinflammation in Parkinson's disease. Neurol Sci 2021; 42:4459-4469. [PMID: 34480241 DOI: 10.1007/s10072-021-05551-1] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 08/07/2021] [Indexed: 02/06/2023]
Abstract
Parkinson's disease (PD) is a chronic and progressive neurodegenerative disorder that affects 1% of the population worldwide. Etiology of PD is likely to be multi-factorial such as protein misfolding, mitochondrial dysfunction, oxidative stress, and neuroinflammation that contributes to the pathology of Parkinson's disease (PD), numerous studies have shown that mitochondrial dysfunction may play a key role in the dopaminergic neuronal loss. In multiple ways, the two most important are the activation of neuroinflammation and mitochondrial dysfunction, while mitochondrial dysfunction could cause neuroinflammation and vice versa. Thus, the mitochondrial proteins are the highly promising target for the development of PD. However, the limited amount of dopaminergic neurons prevented the detailed investigation of Parkinson's disease with regard to mitochondrial dysfunction. Both genetic and environmental factors are also associated with mitochondrial dysfunction and PD pathogenesis. The induction of PD by neurotoxins that inhibit mitochondrial complex I provide direct evidence linking mitochondrial dysfunction to PD. A decrease of mitochondrial complex I activity is observed in PD brain and in neurotoxin- or genetic factor-induced in vitro and in vivo models. Moreover, PINK1, Parkin, DJ-1 and LRRK2 mitochondrial PD gene products have important roles in mitophagy, a cellular process that clear damaged mitochondria. This review paper would discuss the evidence for the mitochondrial dysfunction and neuroinflammation in PD.
Collapse
Affiliation(s)
- Sugumar Mani
- Research and Development Centre, Bharathiar University, Coimbatore, Tamil Nadu, 641046, India
| | - Murugan Sevanan
- Department of Biotechnology, Karunya Institute of Technology and Sciences, Karunya Nagar, Coimbatore, Tamil Nadu, 641114, India.
| | | | - Sathiya Sekar
- Department of Biotechnology, Dr.M.G.R Educational Research Institute, Chennai, India
| |
Collapse
|
75
|
Carmichael K, Sullivan B, Lopez E, Sun L, Cai H. Diverse midbrain dopaminergic neuron subtypes and implications for complex clinical symptoms of Parkinson's disease. AGEING AND NEURODEGENERATIVE DISEASES 2021; 1. [PMID: 34532720 PMCID: PMC8442626 DOI: 10.20517/and.2021.07] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Parkinson’s disease (PD), the most common degenerative movement disorder, is clinically manifested with various motor and non-motor symptoms. Degeneration of midbrain substantia nigra pas compacta (SNc) dopaminergic neurons (DANs) is generally attributed to the motor syndrome. The underlying neuronal mechanisms of non-motor syndrome are largely unexplored. Besides SNc, midbrain ventral tegmental area (VTA) DANs also produce and release dopamine and modulate movement, reward, motivation, and memory. Degeneration of VTA DANs also occurs in postmortem brains of PD patients, implying an involvement of VTA DANs in PD-associated non-motor symptoms. However, it remains to be established that there is a distinct segregation of different SNc and VTA DAN subtypes in regulating different motor and non-motor functions, and that different DAN subpopulations are differentially affected by normal ageing or PD. Traditionally, the distinction among different DAN subtypes was mainly based on the location of cell bodies and axon terminals. With the recent advance of single cell RNA sequencing technology, DANs can be readily classified based on unique gene expression profiles. A combination of specific anatomic and molecular markers shows great promise to facilitate the identification of DAN subpopulations corresponding to different behavior modules under normal and disease conditions. In this review, we first summarize the recent progress in characterizing genetically, anatomically, and functionally diverse midbrain DAN subtypes. Then, we provide perspectives on how the preclinical research on the connectivity and functionality of DAN subpopulations improves our current understanding of cell-type and circuit specific mechanisms of the disease, which could be critically informative for designing new mechanistic treatments.
Collapse
Affiliation(s)
- Kathleen Carmichael
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA.,The Graduate Partnership Program of NIH and Brown University, National Institutes of Health, Bethesda, MD 20892, USA
| | - Breanna Sullivan
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Elena Lopez
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Lixin Sun
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| | - Huaibin Cai
- Transgenic Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
76
|
All Roads Lead to Rome: Different Molecular Players Converge to Common Toxic Pathways in Neurodegeneration. Cells 2021; 10:cells10092438. [PMID: 34572087 PMCID: PMC8468417 DOI: 10.3390/cells10092438] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Revised: 09/12/2021] [Accepted: 09/14/2021] [Indexed: 12/14/2022] Open
Abstract
Multiple neurodegenerative diseases (NDDs) such as Alzheimer’s disease (AD), Parkinson’s disease (PD), amyotrophic lateral sclerosis (ALS) and Huntington’s disease (HD) are being suggested to have common cellular and molecular pathological mechanisms, characterized mainly by protein misfolding and aggregation. These large inclusions, most likely, represent an end stage of a molecular cascade; however, the soluble misfolded proteins, which take part in earlier steps of this cascade, are the more toxic players. These pathological proteins, which characterize each specific disease, lead to the selective vulnerability of different neurons, likely resulting from a combination of different intracellular mechanisms, including mitochondrial dysfunction, ER stress, proteasome inhibition, excitotoxicity, oxidative damage, defects in nucleocytoplasmic transport, defective axonal transport and neuroinflammation. Damage within these neurons is enhanced by damage from the nonneuronal cells, via inflammatory processes that accelerate the progression of these diseases. In this review, while acknowledging the hallmark proteins which characterize the most common NDDs; we place specific focus on the common overlapping mechanisms leading to disease pathology despite these different molecular players and discuss how this convergence may occur, with the ultimate hope that therapies effective in one disease may successfully translate to another.
Collapse
|
77
|
Qi JH, Dong FX. The relevant targets of anti-oxidative stress: a review. J Drug Target 2021; 29:677-686. [PMID: 33401976 DOI: 10.1080/1061186x.2020.1870987] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 12/28/2020] [Indexed: 12/29/2022]
Abstract
Previous studies have found that oxidative stress is the negative reaction of the imbalance between oxidation and antioxidation caused by free radicals, and it is the fuse of aging and many diseases. Scavenging the accumulation of free radicals in the body and inhibiting the production of free radicals are effective ways to reduce the occurrence of oxidative stress. In recent years, studies have found that oxidative stress has other effects on the body, such as anti-tumour. In this paper, the targets related to anti-oxidative stress were introduced, and they were divided into nuclear transcription factors, enzymes, solute carrier family 7, member 11 (SLC7A11) genes and iron death, ion channels, molecular chaperones, small molecules according to their different functions. In addition, we introduce the research status of agonists/inhibitors related to these targets, so as to provide some reference for the follow-up research and clinical application of anti-oxidative stress drugs.
Collapse
Affiliation(s)
- Jian-Hong Qi
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fang-Xu Dong
- College of Foreign Languages, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
78
|
Pan D, Machado L, Bica CG, Machado AK, Steffani JA, Cadoná FC. In Vitro Evaluation of Antioxidant and Anticancer Activity of Lemongrass ( Cymbopogon citratus (D.C.) Stapf). Nutr Cancer 2021; 74:1474-1488. [PMID: 34282694 DOI: 10.1080/01635581.2021.1952456] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Cancer is considered a multifactorial disease and its development could be associated with several factors, for example, rotenone exposition. Unfortunately, many cancers are resistant to chemotherapy, as cervical cancer. Regarding this, lemongrass is a remarkable natural product that presents antioxidant and anticancer activities, which could show therapeutic action against rotenone and cervical cancer. Thus, this study aimed to investigate the antioxidant and anticancer action of lemongrass. An in vitro study was conducted using VERO (kidney cells) and SiHa cell lines (cervical cancer cells). VERO cells were exposed to rotenone and lemongrass extract for 24 and 72 h. While SiHa cells were exposed to lemongrass isolated and associated to chemotherapy, 5-fluorouracil, during 24 and 48 h. After, levels of viability, proliferation, and oxidative metabolism were determined. The results showed that lemongrass presents antioxidant activity on VERO cells by increasing cell viability and proliferation and decreasing oxidative stress caused by rotenone. Moreover, lemongrass showed anticancer activity by decreasing cell viability and increasing oxidative stress parameters on SiHa. Besides, lemongrass had no alteration in the chemotherapy activity. Therefore, this study revealed that lemongrass presents antioxidant and anticancer activity since it can protect against the cytotoxicity of rotenone and reduce the cell viability of cervical cancer.
Collapse
Affiliation(s)
- Daiane Pan
- Health Sciences, University of West Santa Catarina, Joaçaba, Brazil
| | - Larissa Machado
- Biological Sciences, University of West Santa Catarina, Joaçaba, Brazil
| | - Claudia Giuliano Bica
- Health Sciences, Federal University of Health Sciences of Porto Alegre, Porto Alegre, Brazil
| | | | | | | |
Collapse
|
79
|
Ganguly U, Singh S, Pal S, Prasad S, Agrawal BK, Saini RV, Chakrabarti S. Alpha-Synuclein as a Biomarker of Parkinson's Disease: Good, but Not Good Enough. Front Aging Neurosci 2021; 13:702639. [PMID: 34305577 PMCID: PMC8298029 DOI: 10.3389/fnagi.2021.702639] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Accepted: 06/07/2021] [Indexed: 12/15/2022] Open
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative disorder of the elderly, presenting primarily with symptoms of motor impairment. The disease is diagnosed most commonly by clinical examination with a great degree of accuracy in specialized centers. However, in some cases, non-classical presentations occur when it may be difficult to distinguish the disease from other types of degenerative or non-degenerative movement disorders with overlapping symptoms. The diagnostic difficulty may also arise in patients at the early stage of PD. Thus, a biomarker could help clinicians circumvent such problems and help them monitor the improvement in disease pathology during anti-parkinsonian drug trials. This review first provides a brief overview of PD, emphasizing, in the process, the important role of α-synuclein in the pathogenesis of the disease. Various attempts made by the researchers to develop imaging, genetic, and various biochemical biomarkers for PD are then briefly reviewed to point out the absence of a definitive biomarker for this disorder. In view of the overwhelming importance of α-synuclein in the pathogenesis, a detailed analysis is then made of various studies to establish the biomarker potential of this protein in PD; these studies measured total α-synuclein, oligomeric, and post-translationally modified forms of α-synuclein in cerebrospinal fluid, blood (plasma, serum, erythrocytes, and circulating neuron-specific extracellular vesicles) and saliva in combination with certain other proteins. Multiple studies also examined the accumulation of α-synuclein in various forms in PD in the neural elements in the gut, submandibular glands, skin, and the retina. The measurements of the levels of certain forms of α-synuclein in some of these body fluids or their components or peripheral tissues hold a significant promise in establishing α-synuclein as a definitive biomarker for PD. However, many methodological issues related to detection and quantification of α-synuclein have to be resolved, and larger cross-sectional and follow-up studies with controls and patients of PD, parkinsonian disorders, and non-parkinsonian movement disorders are to be undertaken.
Collapse
Affiliation(s)
- Upasana Ganguly
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Sukhpal Singh
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Soumya Pal
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Suvarna Prasad
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Bimal K. Agrawal
- Department of General Medicine, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Reena V. Saini
- Department of Biotechnology, Maharishi Markandeshwar Engineering College, Maharishi Markandeshwar Deemed University, Ambala, India
| | - Sasanka Chakrabarti
- Department of Biochemistry and Central Research Laboratory, Maharishi Markandeshwar Institute of Medical Sciences and Research, Maharishi Markandeshwar Deemed University, Ambala, India
| |
Collapse
|
80
|
Anandhan A, Nguyen N, Syal A, Dreher LA, Dodson M, Zhang DD, Madhavan L. NRF2 Loss Accentuates Parkinsonian Pathology and Behavioral Dysfunction in Human α-Synuclein Overexpressing Mice. Aging Dis 2021; 12:964-982. [PMID: 34221542 PMCID: PMC8219498 DOI: 10.14336/ad.2021.0511] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Accepted: 05/11/2021] [Indexed: 12/14/2022] Open
Abstract
Nuclear factor (erythroid-derived 2)-like 2 (NRF2) is a central regulator of cellular stress responses and its transcriptional activation promotes multiple cellular defense and survival mechanisms. The loss of NRF2 has been shown to increase oxidative and proteotoxic stress, two key pathological features of neurodegenerative disorders such as Parkinson's disease (PD). Moreover, compromised redox homeostasis and protein quality control can cause the accumulation of pathogenic proteins, including alpha-synuclein (α-Syn) which plays a key role in PD. However, despite this link, the precise mechanisms by which NRF2 may regulate PD pathology is not clear. In this study, we generated a humanized mouse model to study the importance of NRF2 in the context of α-Syn-driven neuropathology in PD. Specifically, we developed NRF2 knockout and wild-type mice that overexpress human α-Syn (hα-Syn+/Nrf2-/- and hα-Syn+/Nrf2+/+ respectively) and tested changes in their behavior through nest building, challenging beam, and open field tests at three months of age. Cellular and molecular alterations in α-Syn, including phosphorylation and subsequent oligomerization, as well as changes in oxidative stress, inflammation, and autophagy were also assessed across multiple brain regions. It was observed that although monomeric α-Syn levels did not change, compared to their wild-type counterparts, hα-Syn+/Nrf2-/- mice exhibited increased phosphorylation and oligomerization of α-Syn. This was associated with a loss of tyrosine hydroxylase expressing dopaminergic neurons in the substantia nigra, and more pronounced behavioral deficits reminiscent of early-stage PD, in the hα-Syn+/Nrf2-/- mice. Furthermore, hα-Syn+/Nrf2-/- mice showed significantly amplified oxidative stress, greater expression of inflammatory markers, and signs of increased autophagic burden, especially in the midbrain, striatum and cortical brain regions. These results support an important role for NRF2, early in PD progression. More broadly, it indicates NRF2 biology as fundamental to PD pathogenesis and suggests that targeting NRF2 activation may delay the onset and progression of PD.
Collapse
Affiliation(s)
- Annadurai Anandhan
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
| | - Nhat Nguyen
- Physiology Undergraduate Program, Tucson, AZ, USA.
| | - Arjun Syal
- Neuroscience and Cognitive Science Undergraduate Program, Tucson, AZ, USA.
| | - Luke A Dreher
- Ecology and Evolutionary Biology Undergraduate Program, Tucson, AZ, USA.
| | - Matthew Dodson
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Donna D Zhang
- Pharmacology and Toxicology, University of Arizona, Tucson, AZ, USA.
| | - Lalitha Madhavan
- Department of Neurology, University of Arizona, Tucson, AZ, USA.
- Evelyn F McKnight Brain Institute and Bio5 Institute, University of Arizona, Tucson, AZ, USA.
| |
Collapse
|
81
|
Yarmohammadi F, Hayes AW, Karimi G. Protective effects of curcumin on chemical and drug-induced cardiotoxicity: a review. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2021; 394:1341-1353. [PMID: 33666716 DOI: 10.1007/s00210-021-02072-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Accepted: 02/18/2021] [Indexed: 12/15/2022]
Abstract
Cardiotoxicity is a major adverse effect that can be induced by both therapeutic agents and industrial chemicals. The pathogenesis of such cardiac damage is multifactorial, often injuring the cardiac tissue by generating free radicals, oxidative stress, and/or inflammation. Curcumin (CUR) is a bright yellow chemical produced by Curcuma longa plants. It is the principal curcuminoid of turmeric (Curcuma longa), a member of the ginger family, Zingiberaceae. Administration of CUR has been reported to ameliorate the chemical and drug-induced cardiac injury in several studies. CUR has been suggested to act as an effective candidate against oxidative stress and inflammation in heart tissue via regulation of Nrf2 and suppression of p38 MAPK/NF-κB and NLRP3 inflammasomes. The anti-apoptotic properties of CUR have also been reported to modulate the AMPK, Akt, JNK, and ERK signaling pathways. This review explores the potential protective effects of CUR regarding the detrimental effects often observed in cardiac tissue following exposure to several chemicals including drugs.
Collapse
Affiliation(s)
- Fatemeh Yarmohammadi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - A Wallace Hayes
- Center for Environmental Occupational Risk Analysis and Management, College of Public Health, University of South Florida, Tampa, FL, 33617, USA
| | - Gholamreza Karimi
- Department of Pharmacodynamics and Toxicology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
- Pharmaceutical Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
82
|
Ray B, Mahalakshmi AM, Tuladhar S, Bhat A, Srinivasan A, Pellegrino C, Kannan A, Bolla SR, Chidambaram SB, Sakharkar MK. "Janus-Faced" α-Synuclein: Role in Parkinson's Disease. Front Cell Dev Biol 2021; 9:673395. [PMID: 34124057 PMCID: PMC8194081 DOI: 10.3389/fcell.2021.673395] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 04/15/2021] [Indexed: 01/03/2023] Open
Abstract
Parkinson's disease (PD) is a pathological condition characterized by the aggregation and the resultant presence of intraneuronal inclusions termed Lewy bodies (LBs) and Lewy neurites which are mainly composed of fibrillar α-synuclein (α-syn) protein. Pathogenic aggregation of α-syn is identified as the major cause of LBs deposition. Several mutations in α-syn showing varied aggregation kinetics in comparison to the wild type (WT) α-syn are reported in PD (A30P, E46K, H 50Q, G51D, A53E, and A53T). Also, the cell-to-cell spread of pathological α-syn plays a significant role in PD development. Interestingly, it has also been suggested that the pathology of PD may begin in the gastrointestinal tract and spread via the vagus nerve (VN) to brain proposing the gut-brain axis of α-syn pathology in PD. Despite multiple efforts, the behavior and functions of this protein in normal and pathological states (specifically in PD) is far from understood. Furthermore, the etiological factors responsible for triggering aggregation of this protein remain elusive. This review is an attempt to collate and present latest information on α-syn in relation to its structure, biochemistry and biophysics of aggregation in PD. Current advances in therapeutic efforts toward clearing the pathogenic α-syn via autophagy/lysosomal flux are also reviewed and reported.
Collapse
Affiliation(s)
- Bipul Ray
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Arehally M. Mahalakshmi
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
| | - Sunanda Tuladhar
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Abid Bhat
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
| | - Asha Srinivasan
- Division of Nanoscience & Technology, Faculty of Life Sciences, JSS Academy of Higher Education & Research, Mysuru, India
| | - Christophe Pellegrino
- Institut National de la Santé et de la Recherche Médicale, Institute of Mediterranean Neurobiology, Aix-Marseille University, Marseille, France
| | - Anbarasu Kannan
- Department of Protein Chemistry and Technology, CSIR-Central Food Technological Research Institute, Mysuru, India
| | - Srinivasa Rao Bolla
- Department of Biomedical Sciences, School of Medicine, Nazarbayev University, Nur-Sultan City, Kazakhstan
| | - Saravana Babu Chidambaram
- Department of Pharmacology, JSS College of Pharmacy, JSS Academy of Higher Education & Research, Mysuru, India
- Centre for Experimental Pharmacology and Toxicology, Central Animal Facility, JSS Academy of Higher Education & Research, Mysuru, India
- Special Interest Group – Brain, Behaviour, and Cognitive Neurosciences Research, JSS Academy of Higher Education & Research, Mysuru, India
| | | |
Collapse
|
83
|
Solana-Manrique C, Muñoz-Soriano V, Sanz FJ, Paricio N. Oxidative modification impairs SERCA activity in Drosophila and human cell models of Parkinson's disease. Biochim Biophys Acta Mol Basis Dis 2021; 1867:166152. [PMID: 33892078 DOI: 10.1016/j.bbadis.2021.166152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 03/10/2021] [Accepted: 04/13/2021] [Indexed: 01/25/2023]
Abstract
DJ-1 is a causative gene for familial Parkinson's disease (PD) with different functions, standing out its role against oxidative stress (OS). Accordingly, PD model flies harboring a mutation in the DJ-1β gene (the Drosophila ortholog of human DJ-1) show high levels of OS markers like protein carbonylation, a common post-translational modification that may alter protein function. To increase our understanding of PD pathogenesis as well as to discover potential therapeutic targets for pharmacological intervention, we performed a redox proteomic assay in DJ-1β mutant flies. Among the proteins that showed increased carbonylation levels in PD model flies, we found SERCA, an endoplasmic reticulum Ca2+ channel that plays an important role in Ca2+ homeostasis. Interestingly, several studies have supported the involvement of Ca2+ dyshomeostasis in PD. Thus, we decided to study the relation between SERCA activity and PD physiopathology. Our results showed that SERCA enzymatic activity is significantly reduced in DJ-1β mutant flies, probably as a consequence of OS-induced carbonylation, as well as in a human cell PD model based on DJ-1-deficiency. Indeed, higher carbonylation levels of SERCA were also observed in DJ-1-deficient cells compared to controls. In addition, the specific activator of SERCA, CDN1163, was also able to restore PD-related phenotypes in both familial PD models by increasing SERCA activity. Taken together, our results indicate that impaired SERCA activity due to oxidative modification may play a role in PD physiopathology. Furthermore, we demonstrate that therapeutic strategies addressing SERCA activation could be beneficial to treat this disease as shown for CDN1163.
Collapse
Affiliation(s)
- Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Verónica Muñoz-Soriano
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100 Burjassot, Spain; Instituto Universitario de Biotecnologia y Biomedicina (BIOTECMED), Universidad de Valencia, 46100 Burjassot, Spain.
| |
Collapse
|
84
|
Kurpik M, Zalewski P, Kujawska M, Ewertowska M, Ignatowicz E, Cielecka-Piontek J, Jodynis-Liebert J. Can Cranberry Juice Protect against Rotenone-Induced Toxicity in Rats? Nutrients 2021; 13:1050. [PMID: 33805023 PMCID: PMC8063919 DOI: 10.3390/nu13041050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Revised: 03/19/2021] [Accepted: 03/20/2021] [Indexed: 12/12/2022] Open
Abstract
The high polyphenols content of cranberry accounts for its strong antioxidant activity underlying the beneficial health effects of this fruit. Rotenone (ROT) is a specific inhibitor of mitochondrial complex I in the brain which leads to the generation of oxidative stress. To date, there are few data indicating that toxicity of ROT is not limited to the brain but can also affect other tissues. We aimed to examine whether ROT-induced oxidative stress could be counteracted by cranberry juice not only in the brain but also in the liver and kidney. Wistar rats were given the combined treatment with ROT and cranberry juice (CJ) for 35 days. Parameters of antioxidant status were determined in the organs. ROT enhanced lipid peroxidation solely in the brain. The increase in the DNA damage was noticed in all organs examined and in leukocytes. The beneficial effect of CJ on these parameters appeared only in the brain. Additionally, CJ decreased the activity of serum hepatic enzymes. The effect of CJ on antioxidant enzymes was not consistent, however, in some organs, CJ reversed changes evoked by ROT. Summing up, ROT can cause oxidative damage not only in the brain but also in other organs. CJ demonstrated a protective effect against ROT-induced toxicity.
Collapse
Affiliation(s)
- Monika Kurpik
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland; (M.K.); (M.E.); (J.J.-L.)
| | - Przemysław Zalewski
- Department of Pharmacognosy, Poznan University of Medical Sciences, Święcickiego 4, 60-781 Poznań, Poland; (P.Z.); (J.C.-P.)
| | - Małgorzata Kujawska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland; (M.K.); (M.E.); (J.J.-L.)
| | - Małgorzata Ewertowska
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland; (M.K.); (M.E.); (J.J.-L.)
| | - Ewa Ignatowicz
- Department of Pharmaceutical Biochemistry, Poznan University of Medical Sciences, ul. Święcickiego 4, 60-781 Poznań, Poland;
| | - Judyta Cielecka-Piontek
- Department of Pharmacognosy, Poznan University of Medical Sciences, Święcickiego 4, 60-781 Poznań, Poland; (P.Z.); (J.C.-P.)
| | - Jadwiga Jodynis-Liebert
- Department of Toxicology, Poznan University of Medical Sciences, Dojazd 30, 60-631 Poznań, Poland; (M.K.); (M.E.); (J.J.-L.)
| |
Collapse
|
85
|
Fernandes EJ, Poetini MR, Barrientos MS, Bortolotto VC, Araujo SM, Santos Musachio EA, De Carvalho AS, Leimann FV, Gonçalves OH, Ramborger BP, Roehrs R, Prigol M, Guerra GP. Exposure to lutein-loaded nanoparticles attenuates Parkinson's model-induced damage in Drosophila melanogaster: Restoration of dopaminergic and cholinergic system and oxidative stress indicators. Chem Biol Interact 2021; 340:109431. [PMID: 33716020 DOI: 10.1016/j.cbi.2021.109431] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/27/2021] [Accepted: 02/27/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's is a neurodegenerative disease, characterized by the loss of dopaminergic neurons, cholinergic alterations and oxidative damages. Lutein is widely known by its antioxidants properties. In the present study, we investigated whether lutein-loaded nanoparticles protects against locomotor damage and neurotoxicity induced by Parkinson's disease model in Drosophila melanogaster, as well as possible mechanisms of action. First, the nanoparticles were characterized by physicochemical methods, demonstrating that water affinity was improved by the encapsulation of lutein into the polymeric encapsulant matrix. The fruit flies of 1-4 days old were divided into four groups and exposed to a standard diet (control), a diet containing either rotenone (500 μM), lutein-loaded nanoparticles (6 μM) or rotenone (500 μM) and lutein-loaded nanoparticles (6 μM) for 7 days. The survival percentage was assessed, the flies were submitted to negative geotaxis, open field tasks and the determination of dopamine levels, tyrosine hydroxylase (TH) and acetylcholinesterase activities and oxidative stress indicators (superoxide dismutase, catalase, thiobarbituric acid reactive substances and glutathione S-transferase) were carried out. The exposure to lutein-loaded nanoparticles protected against locomotor damage and the decrease survival rate induced by rotenone, besides, it restored the dopamine levels, TH and acetylcholinesterase activities and oxidative stress indicators. These results provide evidence that lutein-loaded nanoparticles are an alternative treatment for rotenone-induced damage, and suggest the involvement of dopaminergic and cholinergic system and oxidative stress.
Collapse
Affiliation(s)
- Eliana Jardim Fernandes
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Marcia Rósula Poetini
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Magna Sotelo Barrientos
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil
| | - Vandreza Cardoso Bortolotto
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Stífani Machado Araujo
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Elize Aparecida Santos Musachio
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Amarilis Santos De Carvalho
- Programa de Pós-Graduação em Tecnologia de Alimentos, Universidade Tecnológica Federal do Paraná - Campus Campo Mourão, 87301-006, Campo Mourão, PR, Brazil
| | - Fernanda Vitória Leimann
- Programa de Pós-Graduação em Tecnologia de Alimentos, Universidade Tecnológica Federal do Paraná - Campus Campo Mourão, 87301-006, Campo Mourão, PR, Brazil
| | - Odinei Hess Gonçalves
- Programa de Pós-Graduação em Tecnologia de Alimentos, Universidade Tecnológica Federal do Paraná - Campus Campo Mourão, 87301-006, Campo Mourão, PR, Brazil
| | - Bruna Piaia Ramborger
- Grupo Interdisciplinar de Pesquisa em Prática de Ensino (GIPPE), Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Rafael Roehrs
- Grupo Interdisciplinar de Pesquisa em Prática de Ensino (GIPPE), Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Marina Prigol
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil
| | - Gustavo Petri Guerra
- Laboratório de Avaliações Farmacológicas e Toxicológicas Aplicadas às Moléculas Bioativas - LaftamBio, Universidade Federal Do Pampa - Campus Itaqui, 97650-000, Itaqui, RS, Brazil; Programa de Pós-Graduação em Bioquímica, Universidade Federal do Pampa - Campus Uruguaiana, 97508-000, Uruguaiana, RS, Brazil.
| |
Collapse
|
86
|
Suppressing Cdk5 Activity by Luteolin Inhibits MPP +-Induced Apoptotic of Neuroblastoma through Erk/Drp1 and Fak/Akt/GSK3β Pathways. Molecules 2021; 26:molecules26051307. [PMID: 33671094 PMCID: PMC7957557 DOI: 10.3390/molecules26051307] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 02/23/2021] [Accepted: 02/23/2021] [Indexed: 02/07/2023] Open
Abstract
Parkinson’s disease (PD) is characterized by the progressive degeneration of dopaminergic neurons. The cause of PD is still unclear. Oxidative stress and mitochondrial dysfunction have been linked to the development of PD. Luteolin, a non-toxic flavonoid, has become interested in an alternative medicine, according to its effects on anti-oxidative stress and anti-apoptosis, although the underlying mechanism of luteolin on PD has not been fully elucidated. This study aims to investigate whether luteolin prevents neurotoxicity induction by 1-methyl-4-phenylpyridinium iodide (MPP+), a neurotoxin in neuroblastoma SH-SY5Y cells. The results reveal that luteolin significantly improved cell viability and reduced apoptosis in MPP+-treated cells. Increasing lipid peroxidation and superoxide anion (O2−), including mitochondrial membrane potential (Δψm) disruption, is ameliorated by luteolin treatment. In addition, luteolin attenuated MPP+-induced neurite damage via GAP43 and synapsin-1. Furthermore, Cdk5 is found to be overactivated and correlated with elevation of cleaved caspase-3 activity in MPP+-exposed cells, while phosphorylation of Erk1/2, Drp1, Fak, Akt and GSK3β are inhibited. In contrast, luteolin attenuated Cdk5 overactivation and supported phosphorylated level of Erk1/2, Drp1, Fak, Akt and GSK3β with reducing in cleaved caspase-3 activity. Results indicate that luteolin exerts neuroprotective effects via Cdk5-mediated Erk1/2/Drp1 and Fak/Akt/GSK3β pathways, possibly representing a potential preventive agent for neuronal disorder.
Collapse
|
87
|
Arab HH, Safar MM, Shahin NN. Targeting ROS-Dependent AKT/GSK-3β/NF-κB and DJ-1/Nrf2 Pathways by Dapagliflozin Attenuates Neuronal Injury and Motor Dysfunction in Rotenone-Induced Parkinson's Disease Rat Model. ACS Chem Neurosci 2021; 12:689-703. [PMID: 33543924 DOI: 10.1021/acschemneuro.0c00722] [Citation(s) in RCA: 110] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Dapagliflozin, a selective sodium-glucose co-transporter 2 (SGLT2) inhibitor, has emerged as a promising neuroprotective agent in murine models of epilepsy and obesity-induced cognitive impairment through its marked antioxidant/antiapoptotic features. However, the impact of dapagliflozin on the pathogenesis of Parkinson's disease (PD) is lacking. Hence, the present study aimed at exploring the potential neuroprotective effects of dapagliflozin against PD-associated neurodegenerative aberrations/motor dysfunction in rotenone-induced PD rat model. Rotenone (1.5 mg/kg) was subcutaneously administered every other day for 3 weeks. The expression of target signals was investigated using qPCR, Western blotting, ELISA, and immunohistochemistry. Dapagliflozin (1 (mg/kg)/day, by gavage for 3 weeks) attenuated PD motor dysfunction and improved motor coordination in the open-field and rotarod tests without triggering hypoglycemia. It also diminished the histopathologic alterations and α-synuclein expression and augmented tyrosine hydroxylase and dopamine levels. Dapagliflozin markedly alleviated neuronal oxidative stress via lowering lipid peroxides with consequent restoration of the disturbed DJ-1/Nrf2 pathway. Moreover, dapagliflozin counteracted ROS-dependent neuronal apoptosis and upregulated GDNF and its downstream PI3K/AKT/GSK-3β (Ser9) pathway. Meanwhile, it suppressed neuroinflammation via curbing the activation of NF-κB pathway and TNF-α levels. Together, these pleiotropic neuroprotective effects highlight the promising role of dapagliflozin in the management of PD.
Collapse
Affiliation(s)
- Hany H. Arab
- Department of Pharmacology and Toxicology, College of Pharmacy, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| | - Marwa M. Safar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt, Cairo 11837, Egypt
| | - Nancy N. Shahin
- Department of Biochemistry, Faculty of Pharmacy, Cairo University, Cairo 11562, Egypt
| |
Collapse
|
88
|
Hor SL, Teoh SL, Lim WL. Plant Polyphenols as Neuroprotective Agents in Parkinson's Disease Targeting Oxidative Stress. Curr Drug Targets 2021; 21:458-476. [PMID: 31625473 DOI: 10.2174/1389450120666191017120505] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 09/26/2019] [Accepted: 09/26/2019] [Indexed: 12/15/2022]
Abstract
Parkinson's disease (PD) is the second most prevalent progressive neurodegenerative disorder characterized by the degeneration of dopaminergic neurons in the human midbrain. Various ongoing research studies are competing to understand the pathology of PD and elucidate the mechanisms underlying neurodegeneration. Current pharmacological treatments primarily focused on improving dopamine metabolism in PD patients, despite the side effects of long-term usage. In recent years, it is recognized that oxidative stress-mediated pathways lead to neurodegeneration in the brain, which is associated with the pathophysiology of PD. The importance of oxidative stress is often less emphasized when developing potential therapeutic approaches. Natural plant antioxidants have been shown to mediate the oxidative stress-induced effects in PD, which has gained considerable attention in both in vitro and in vivo studies. Yet, clinical trials on natural polyphenol compounds are limited, restricting the potential use of these compounds as an alternative treatment for PD. Therefore, this review provides an understanding of the oxidative stress-induced effects in PD by elucidating the underlying events contributing to oxidative stress and explore the potential use of polyphenols in improving the oxidative status in PD. Preclinical findings have supported the potential of polyphenols in providing neuroprotection against oxidative stress-induced toxicity in PD. However, limiting factors, such as safety and bioavailability of polyphenols, warrant further investigations so as to make them the potential target for clinical applications in the treatment and management of PD.
Collapse
Affiliation(s)
- Suet Lee Hor
- Department of Biological Sciences, School of Science and Technology, Sunway University, 47500 Selangor, Malaysia
| | - Seong Lin Teoh
- Department of Anatomy, Universiti Kebangsaan Malaysia Medical Centre, 56000 Kuala Lumpur, Malaysia
| | - Wei Ling Lim
- Department of Biological Sciences, School of Science and Technology, Sunway University, 47500 Selangor, Malaysia
| |
Collapse
|
89
|
Mina PR, Kumar S, Agarwal K, Kumar R, Pal A, Tandon S, Yadav SK, Yadav S, Darokar MP. 4-chloro eugenol interacts synergistically with artesunate against drug resistant P. falciparum inducing oxidative stress. Biomed Pharmacother 2021; 137:111311. [PMID: 33524782 DOI: 10.1016/j.biopha.2021.111311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 01/10/2021] [Accepted: 01/21/2021] [Indexed: 11/29/2022] Open
Abstract
4-chloro eugenol (4CE), a semisynthetic analog of phytomolecule eugenol exhibited potent antiplasmodial activity with IC50 in the range of 1.5-5 μM against sensitive as well as drug resistant strain of P. falciparum. This analog also showed synergy with a clinically used antimalarial drug artesunate and was able to curtail the IC50 of artesunate up to 4-5 folds. Although, 4CE did not show any effect on heme polymerization, the most common drug target in the malaria parasite, it could increase the level of reactive oxygen species (ROS) and reactive nitrogen species (RNS) alone as well as in combination with artesunate. Further, 4CE induced oxidative stress was observed to affect the macromolecules in terms of DNA damage, protein carbonylation and lipid peroxidation. At the physiological level, cellular organelles like mitochondria and endoplasmic reticulum were observed to be get affected by 4CE in terms of membrane depolarization and calcium ion leakage respectively. These observations could be validated by expression analysis of oxidative stress responsive genes and proteins. Further, in in vivo assay, 4CE showed significant chemo-suppression of parasitemia as well as an increase in mean survival time in the murine malaria model. Interestingly, in combination with artesunate, 4CE showed higher chemo-suppression as well as enhanced mean survival time at a much lower concentrations of both the partners as compared to an individual dose of artesunate and 4CE. A combination of 4CE and artesunate was also observed to attenuate cerebral malaria pathogenesis.
Collapse
Affiliation(s)
- Pooja Rani Mina
- Bioprospectionand Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Saurabh Kumar
- Bioprospectionand Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Karishma Agarwal
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Ravi Kumar
- Bioprospectionand Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Anirban Pal
- Bioprospectionand Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Sudeep Tandon
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Sanjeev Kumar Yadav
- Developmental Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Sanjay Yadav
- Developmental Toxicology Laboratory, CSIR-Indian Institute of Toxicology Research, Lucknow, India
| | - Mahendra P Darokar
- Bioprospectionand Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India.
| |
Collapse
|
90
|
Gonzalez-Hunt CP, Luz AL, Ryde IT, Turner EA, Ilkayeva OR, Bhatt DP, Hirschey MD, Meyer JN. Multiple metabolic changes mediate the response of Caenorhabditis elegans to the complex I inhibitor rotenone. Toxicology 2021; 447:152630. [PMID: 33188857 PMCID: PMC7750303 DOI: 10.1016/j.tox.2020.152630] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2020] [Revised: 11/05/2020] [Accepted: 11/09/2020] [Indexed: 12/21/2022]
Abstract
Rotenone, a mitochondrial complex I inhibitor, has been widely used to study the effects of mitochondrial dysfunction on dopaminergic neurons in the context of Parkinson's disease. Although the deleterious effects of rotenone are well documented, we found that young adult Caenorhabditis elegans showed resistance to 24 and 48 h rotenone exposures. To better understand the response to rotenone in C. elegans, we evaluated mitochondrial bioenergetic parameters after 24 and 48 h exposures to 1 μM or 5 μM rotenone. Results suggested upregulation of mitochondrial complexes II and V following rotenone exposure, without major changes in oxygen consumption or steady-state ATP levels after rotenone treatment at the tested concentrations. We found evidence that the glyoxylate pathway (an alternate pathway not present in higher metazoans) was induced by rotenone exposure; gene expression measurements showed increases in mRNA levels for two complex II subunits and for isocitrate lyase, the key glyoxylate pathway enzyme. Targeted metabolomics analyses showed alterations in the levels of organic acids, amino acids, and acylcarnitines, consistent with the metabolic restructuring of cellular bioenergetic pathways including activation of complex II, the glyoxylate pathway, glycolysis, and fatty acid oxidation. This expanded understanding of how C. elegans responds metabolically to complex I inhibition via multiple bioenergetic adaptations, including the glyoxylate pathway, will be useful in interrogating the effects of mitochondrial and bioenergetic stressors and toxicants.
Collapse
Affiliation(s)
- Claudia P Gonzalez-Hunt
- Department of Nicholas School of the Environment, Duke University, Durham, NC, 27708, United States
| | - Anthony L Luz
- Department of Nicholas School of the Environment, Duke University, Durham, NC, 27708, United States
| | - Ian T Ryde
- Department of Nicholas School of the Environment, Duke University, Durham, NC, 27708, United States
| | - Elena A Turner
- Department of Nicholas School of the Environment, Duke University, Durham, NC, 27708, United States
| | - Olga R Ilkayeva
- Duke Molecular Physiology Institute, Durham, NC, 27710, United States; Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, 27710, United States
| | - Dhaval P Bhatt
- Duke Molecular Physiology Institute, Durham, NC, 27710, United States
| | - Matthew D Hirschey
- Duke Molecular Physiology Institute, Durham, NC, 27710, United States; Sarah W. Stedman Nutrition and Metabolism Center, Durham, NC, 27710, United States; Departments of Medicine and Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC, 27710, United States
| | - Joel N Meyer
- Department of Nicholas School of the Environment, Duke University, Durham, NC, 27708, United States.
| |
Collapse
|
91
|
Rottenberg H, Hoek JB. The Mitochondrial Permeability Transition: Nexus of Aging, Disease and Longevity. Cells 2021; 10:cells10010079. [PMID: 33418876 PMCID: PMC7825081 DOI: 10.3390/cells10010079] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 12/23/2020] [Accepted: 01/01/2021] [Indexed: 12/11/2022] Open
Abstract
The activity of the mitochondrial permeability transition pore, mPTP, a highly regulated multi-component mega-channel, is enhanced in aging and in aging-driven degenerative diseases. mPTP activity accelerates aging by releasing large amounts of cell-damaging reactive oxygen species, Ca2+ and NAD+. The various pathways that control the channel activity, directly or indirectly, can therefore either inhibit or accelerate aging or retard or enhance the progression of aging-driven degenerative diseases and determine lifespan and healthspan. Autophagy, a catabolic process that removes and digests damaged proteins and organelles, protects the cell against aging and disease. However, the protective effect of autophagy depends on mTORC2/SKG1 inhibition of mPTP. Autophagy is inhibited in aging cells. Mitophagy, a specialized form of autophagy, which retards aging by removing mitochondrial fragments with activated mPTP, is also inhibited in aging cells, and this inhibition leads to increased mPTP activation, which is a major contributor to neurodegenerative diseases, such as Alzheimer's and Parkinson's diseases. The increased activity of mPTP in aging turns autophagy/mitophagy into a destructive process leading to cell aging and death. Several drugs and lifestyle modifications that enhance healthspan and lifespan enhance autophagy and inhibit the activation of mPTP. Therefore, elucidating the intricate connections between pathways that activate and inhibit mPTP, in the context of aging and degenerative diseases, could enhance the discovery of new drugs and lifestyle modifications that slow aging and degenerative disease.
Collapse
Affiliation(s)
- Hagai Rottenberg
- New Hope Biomedical R&D, 23 W. Bridge street, New Hope, PA 18938, USA
- Correspondence: ; Tel.: +1-267-614-5588
| | - Jan B. Hoek
- MitoCare Center, Department of Anatomy, Pathology and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA;
| |
Collapse
|
92
|
Liu X, Qu L, Zhang N, Yu X, Xiao Z, Song L, Xie J, Xu H. Ndfip1 Prevents Rotenone-Induced Neurotoxicity and Upregulation of α-Synuclein in SH-SY5Y Cells. Front Mol Neurosci 2021; 13:613404. [PMID: 33469419 PMCID: PMC7813998 DOI: 10.3389/fnmol.2020.613404] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Accepted: 12/02/2020] [Indexed: 12/21/2022] Open
Abstract
Nedd4 family interacting protein 1 (Ndfip1) is an adaptor of Nedd4-family ubiquitin ligases. Experimental results showed that Ndfip1 had a potential neuroprotective effect in neurology diseases. However, the neuroprotective effect and the underlying mechanisms of Ndfip1 in Parkinson's disease (PD) have not yet been fully elucidated. Therefore, in this study, we explored the neuroprotective effect of Ndfip1 against mitochondrial complex I inhibitor rotenone in a human dopaminergic neuroblastoma SH-SY5Y cell line and further elucidated its possible underlying mechanisms. Our results showed that rotenone could induce the up-regulation of α-synuclein (α-syn) in both mRNA and protein levels. The expression of Ndfip1 decreased at 24 h after rotenone treatment. Further study showed that high expression of Ndfip1 could protect SH-SY5Y cells against rotenone-induced neurotoxicity and antagonize the rotenone-induced increase in α-syn protein levels. In addition, high expression of Ndfip1 inhibited rotenone-induced increase in the protein levels of caspase-3 and decrease in tyrosine hydroxylase (TH). Further study showed that Ndfip1 did not affect the protein expression of iron regulatory protein 1 (IRP1), transferrin receptor 1 (TfR1), while antagonized the increase in protein levels of P62 and ferritin L caused by rotenone. Our findings provide specific identification of Ndfip1 proteins to inhibit the increase of α-syn in rotenone-induced SH-SY5Y cells. Ndfip1 might be a new theoretical drug target for the prevention and treatment of PD.
Collapse
Affiliation(s)
- Xin Liu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China
| | - Le Qu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Na Zhang
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Xiaoqi Yu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Zhixin Xiao
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China
| | - Limei Song
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China
| | - Junxia Xie
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| | - Huamin Xu
- Shandong Provincial Key Laboratory of Pathogenesis and Prevention of Neurological Disorders and State Key Disciplines: Physiology, Department of Physiology, Medical College of Qingdao University, Qingdao, China.,Institute of Brain Science and Disease, Qingdao University, Qingdao, China
| |
Collapse
|
93
|
Thadathil N, Xiao J, Hori R, Alway SE, Khan MM. Brain Selective Estrogen Treatment Protects Dopaminergic Neurons and Preserves Behavioral Function in MPTP-induced Mouse Model of Parkinson's Disease. J Neuroimmune Pharmacol 2020; 16:667-678. [PMID: 33221984 DOI: 10.1007/s11481-020-09972-1] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Accepted: 11/16/2020] [Indexed: 12/20/2022]
Abstract
Parkinson's disease (PD) is characterized by progressive degeneration of dopaminergic neurons in the substantia nigra and loss of both motor and non-motor features. Several clinical and preclinical studies have provided evidence that estrogen therapy reduces the risk of PD but have limitations in terms of adverse peripheral effects. Therefore, we examined the potential beneficial effects of the brain-selective estrogen prodrug, 10β, 17β-dihydroxyestra-1,4-dien-3-one (DHED) on nigrostriatal dopaminergic neurodegeneration and behavioral abnormalities in 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) mouse model of PD. Wild-type mice were treated with daily subcutaneous injections of DHED (50 and 100 µg/kg) or vehicle for four weeks. To produce PD-like symptoms, mice were injected with MPTP (18 mg/kg in saline; intraperitoneally) four times at 2-hr intervals for one day. After behavioral examination, mice were sacrificed, and the brains were isolated for neurochemical and morphological examinations. MPTP injected mice exhibited loss of dopaminergic neurons and fibers in substantia nigra and striatum respectively, along with impaired motor function at day 7 post MPTP injection. These phenotypes were associated with significantly increased oxidative stress and inflammatory responses in the striatum regions. DHED treatments significantly mitigated behavioral impairments and dopaminergic neurodegeneration induced by MPTP. We further observed that DHED treatment suppressed oxidative stress and inflammation in the striatum of MPTP treated mice when compared to vehicle treated mice. In conclusions, our findings suggest that DHED protects dopaminergic neurons from MPTP toxicity in mouse model of PD and support a beneficial effect of brain-selective estrogen in attenuating neurodegeneration and motor symptoms in PD-related neurological disorders. Graphical Abstract.
Collapse
Affiliation(s)
- Nidheesh Thadathil
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Jianfeng Xiao
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA
| | - Roderick Hori
- Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Stephen E Alway
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, 855 Monroe Avenue, 415 Link Building, Memphis, TN, 38163, USA.
- Center for Muscle, Metabolism and Neuropathology, Division of Rehabilitation Sciences, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA.
| |
Collapse
|
94
|
Momtaz AZ, Ahumada Sabagh AD, Gonzalez Amortegui JG, Salazar SA, Finessi A, Hernandez J, Christensen S, Serbus LR. A Role for Maternal Factors in Suppressing Cytoplasmic Incompatibility. Front Microbiol 2020; 11:576844. [PMID: 33240234 PMCID: PMC7680759 DOI: 10.3389/fmicb.2020.576844] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/20/2020] [Indexed: 12/02/2022] Open
Abstract
Wolbachia are maternally transmitted bacterial endosymbionts, carried by approximately half of all insect species. Wolbachia prevalence in nature stems from manipulation of host reproduction to favor the success of infected females. The best known reproductive modification induced by Wolbachia is referred to as sperm-egg Cytoplasmic Incompatibility (CI). In CI, the sperm of Wolbachia-infected males cause embryonic lethality, attributed to paternal chromatin segregation defects during early mitotic divisions. Remarkably, the embryos of Wolbachia-infected females “rescue” CI lethality, yielding egg hatch rates equivalent to uninfected female crosses. Several models have been discussed as the basis for Rescue, and functional evidence indicates a major contribution by Wolbachia CI factors. A role for host contributions to Rescue remains largely untested. In this study, we used a chemical feeding approach to test for CI suppression capabilities by Drosophila simulans. We found that uninfected females exhibited significantly higher CI egg hatch rates in response to seven chemical treatments that affect DNA integrity, cell cycle control, and protein turnover. Three of these treatments suppressed CI induced by endogenous wRi Wolbachia, as well as an ectopic wMel Wolbachia infection. The results implicate DNA integrity as a focal aspect of CI suppression for different Wolbachia strains. The framework presented here, applied to diverse CI models, will further enrich our understanding of host reproductive manipulation by insect endosymbionts.
Collapse
Affiliation(s)
- Ajm Zehadee Momtaz
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Abraham D Ahumada Sabagh
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Julian G Gonzalez Amortegui
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Samuel A Salazar
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Andrea Finessi
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Jethel Hernandez
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Steen Christensen
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| | - Laura R Serbus
- Department of Biological Sciences, Florida International University, Miami, FL, United States.,Biomolecular Sciences Institute, Florida International University, Miami, FL, United States
| |
Collapse
|
95
|
Molecular Mechanism of Platelet-Derived Growth Factor (PDGF)-BB-Mediated Protection Against MPP+ Toxicity in SH-SY5Y Cells. J Mol Neurosci 2020; 71:1131-1143. [DOI: 10.1007/s12031-020-01735-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 10/02/2020] [Indexed: 12/21/2022]
|
96
|
Ardah MT, Bharathan G, Kitada T, Haque ME. Ellagic Acid Prevents Dopamine Neuron Degeneration from Oxidative Stress and Neuroinflammation in MPTP Model of Parkinson's Disease. Biomolecules 2020; 10:E1519. [PMID: 33172035 PMCID: PMC7694688 DOI: 10.3390/biom10111519] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 10/30/2020] [Accepted: 11/04/2020] [Indexed: 02/07/2023] Open
Abstract
Parkinson's disease (PD) is one of the most common neurodegenerative diseases and is characterized by progressive dopaminergic neurodegeneration in the substantia nigra pars compacta area. In the present study, treatment of EA for 1 week at a dose of 10 mg/kg body weight prior to MPTP (25 mg/kg body weight) was carried out. MPTP administration caused oxidative stress, as evidenced by decreased activities of superoxide dismutase and catalase, and the depletion of reduced glutathione with a concomitant rise in the lipid peroxidation product, malondialdehyde. It also significantly increased the pro-inflammatory cytokines and elevated the inflammatory mediators like cyclooxygenase-2 (COX-2) and inducible nitric oxide synthase (iNOS) in the striatum. Immunohistochemical analysis revealed a loss of dopamine neurons in the SNc area and a decrease in dopamine transporter in the striatum following MPTP administration. However, treatment with EA prior to MPTP injection significantly rescued the dopaminergic neurons and dopamine transporter. EA treatment further restored antioxidant enzymes, prevented the depletion of glutathione and inhibited lipid peroxidation, in addition to the attenuation of pro-inflammatory cytokines. EA also reduced the levels of COX-2 and iNOS. The findings of the present study demonstrate that EA protects against MPTP-induced PD and the observed neuroprotective effects can be attributed to its potent antioxidant and anti-inflammatory properties.
Collapse
Affiliation(s)
- Mustafa T. Ardah
- Department of Biochemistry, College of Medicine and Health Sciences, UAEU, Al Ain, UAE; (M.T.A.); (G.B.)
| | - Greeshma Bharathan
- Department of Biochemistry, College of Medicine and Health Sciences, UAEU, Al Ain, UAE; (M.T.A.); (G.B.)
| | - Tohru Kitada
- Otawa-Kagaku Service, Parkinson’s Clinic and Research, Kamakura 247-0061, Japan;
| | - M. Emdadul Haque
- Department of Biochemistry, College of Medicine and Health Sciences, UAEU, Al Ain, UAE; (M.T.A.); (G.B.)
| |
Collapse
|
97
|
Mohammed NA, Abdou HM, Tass MA, Alfwuaires M, Abdel-Moneim AM, Essawy AE. Oral Supplements of <i>Ginkgo biloba</i> Extract Alleviate Neuroinflammation, Oxidative Impairments and Neurotoxicity in Rotenone-Induced Parkinsonian Rats. Curr Pharm Biotechnol 2020; 21:1259-1268. [PMID: 32196446 DOI: 10.2174/1389201021666200320135849] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 03/02/2020] [Accepted: 03/04/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Ginkgo biloba extract (GbE) is known to contain several bioactive compounds and exhibits free radical scavenging activity. Parkinson's Disease (PD) is a neurodegenerative disorder characterized by the loss of dopaminergic neurons and is associated with oxidative stress, neuroinflammation and apoptosis. OBJECTIVE The current study aimed to investigate the neuroprotective effect of GbE in a rat model of PD induced by rotenone (ROT; a neurotoxin). METHODS Twenty-four male albino rats were randomly divided into four groups of six rats each: normal control, GbE treated, toxin control (ROT treated) and GbE+ROT group. RESULTS Oral administration of ROT (2.5 mg/kg b.w.) for 50 days caused an increased generation of lipid peroxidation products and significant depletion of reduced glutathione, total thiol content and activities of enzymatic antioxidants, i.e., superoxide dismutase and glutathione peroxidase in the brains of treated rats. Furthermore, ROT caused an elevation in acetylcholinesterase, interleukin-1β, interleukin- 6 and tumor necrosis factor-α and a significant reduction in dopamine in the stratum and substantia nigra. Immunohistochemical results illustrated that ROT treatment reduced the expression of tyrosine hydroxylase (TH). GbE treatment (150 mg/kg b.w./day) significantly reduced the elevated oxidative stress markers and proinflammatory cytokines and restored the reduced antioxidant enzyme activities, DA level and TH expression. These results were confirmed by histological observations that clearly indicated a neuroprotective effect of GbE against ROT-induced PD. CONCLUSION GbE mitigated ROT-induced PD via the inhibition of free-radical production, scavenging of ROS, and antioxidant enhancement.
Collapse
Affiliation(s)
- Nema A Mohammed
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Heba M Abdou
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| | - Mona A Tass
- Faculty of Art and Science- Badr, Al-Jabal Al-Gharbi University, Gherian, Libya
| | - Manal Alfwuaires
- Department of Biological Sciences, Faculty of Science, King Faisal University, Hofuf-31982, Al-Ahsa, Saudi Arabia
| | | | - Amina E Essawy
- Department of Zoology, Faculty of Science, Alexandria University, Alexandria, Egypt
| |
Collapse
|
98
|
Balan DJ, Rajavel T, Das M, Sathya S, Jeyakumar M, Devi KP. Thymol induces mitochondrial pathway-mediated apoptosis via ROS generation, macromolecular damage and SOD diminution in A549 cells. Pharmacol Rep 2020; 73:240-254. [PMID: 33095436 DOI: 10.1007/s43440-020-00171-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Revised: 10/01/2020] [Accepted: 10/07/2020] [Indexed: 12/15/2022]
Abstract
BACKGROUND Thymol is a monoterpene phenol found in thyme species plants. The present study was carried out to investigate the effect of thymol and its molecular mechanism on non-small lung cancer (A549) cells. METHODS The cytotoxic effect of thymol on A549 cells was assessed via MTT assay. ROS production, macromolecular damage, apoptosis were determined using DCF-DA, PI, AO/EtBr stains, respectively. ROS-dependent effect of thymol was confirmed using NAC. The expression of caspase-9, Bcl-2, Bax and cell cycle profile was analyzed via western blot and FACS, respectively. RESULTS The antiproliferative effect of thymol on A549 cells was found to be both dose and time dependent with IC50 values of 112 μg/ml (745 μM) at 24 h. Thymol treatment favored apoptotic cell death and caused G0/G1 cell cycle arrest. It mediated cellular and nuclear morphological changes, phosphatidylserine translocation, and mitochondrial membrane depolarization. Additionally, upregulation of Bax, downregulation of Bcl-2, and apoptotic fragmented DNA were also observed. Thymol induced ROS by reducing the SOD level which was confirmed via in vitro and in silico analysis. Furthermore, the levels of lipid peroxides and protein carbonyl content were elevated in thymol-treated groups. Notably, N-acetyl cysteine pretreatment reversed the efficacy of thymol on A549 cells. Moreover, thymol-treated human PBMC cells did not show any significant cytotoxicity. CONCLUSION Overall, our results confirmed that thymol can act as a safe and potent therapeutic agent to treat NSCLC.
Collapse
Affiliation(s)
- Devasahayam Jaya Balan
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India
| | - Tamilselvam Rajavel
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India.,Department of Biochemistry and Molecular Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Mamali Das
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India
| | - Sethuraman Sathya
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India
| | - Mahalingam Jeyakumar
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India
| | - Kasi Pandima Devi
- Department of Biotechnology, Alagappa University [Science Campus], Karaikudi, Tamil Nadu, 630 003, India.
| |
Collapse
|
99
|
Pingale T, Gupta GL. Classic and evolving animal models in Parkinson's disease. Pharmacol Biochem Behav 2020; 199:173060. [PMID: 33091373 DOI: 10.1016/j.pbb.2020.173060] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 10/10/2020] [Accepted: 10/12/2020] [Indexed: 02/07/2023]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with motor and non-motor symptoms. PD is characterized by the degeneration of dopaminergic neurons in the substantia nigra pars compacta (SNpc) and deficiency of dopamine in the striatal region. The primary objective in PD research is to understand the pathogenesis, targets, and development of therapeutic interventions to control the progress of the disease. The anatomical and physiological resemblances between humans and animals gathered the researcher's attention towards the use of animals in PD research. Due to varying age of onset, symptoms, and progression rate, PD becomes heterogeneous which demands the variety of animal models to study diverse features of the disease. Parkinson is a multifactorial disorder, selection of models become important as not a single model shows all the biochemical features of the disease. Currently, conventional pharmacological, neurotoxin-induced, genetically modified and cellular models are available for PD research, but none of them recapitulate all the biochemical characteristics of the disease. In this review, we included the updated knowledge on the main features of currently available in vivo and in vitro models as well as their strengths and weaknesses.
Collapse
Affiliation(s)
- Tanvi Pingale
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India
| | - Girdhari Lal Gupta
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM'S NMIMS, V.L. Mehta Road, Vile Parle (W), Mumbai 400 056, India; School of Pharmacy & Technology Management, SVKM'S NMIMS, Shirpur, Maharashtra, India.
| |
Collapse
|
100
|
Thanalakshmi J, Archana R, Senthilkumar S, Shakila R, Pazhanivel N, Subhashini S. Role of caloric vestibular stimulation in improvement of motor symptoms and inhibition of neuronal degeneration in rotenone model of Parkinson's disease - An experimental study. Physiol Int 2020; 107:390-405. [PMID: 33021954 DOI: 10.1556/2060.2020.00036] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 07/27/2020] [Indexed: 11/19/2022]
Abstract
Objective Parkinson's disease (PD) is a progressive neurodegenerative disorder. In order to explore a noninvasive treatment of PD, in the current study the authors evaluated the neuroprotective efficacy of caloric vestibular stimulation (CVS) using the rotenone-induced rat model of PD. The rotenone models of PD are gaining attention due to high reproducibility. It is also considered to be an improved model to exhibit the pathogenesis of PD and test the neuroprotective effect of various therapeutic interventions. Materials and methods Rotenone was i.p. injected (3 mg/kg body weight) to male Wistar albino rats for 21 days to induce PD. As PD is chronic and progressive in nature, the efficacy of chronic CVS intervention was evaluated for 30 days after inducing PD in rats. Motor symptoms were evaluated by assessing locomotor activity in actophotometer, whereas movement analysis was done using Ludolph test and motor coordination was evaluated using rotarod apparatus. The neurochemical and neuropathological changes were also observed in the corpus striatum of rats. Results Rotenone administration showed decreased locomotor activity, motor coordination and general movement associated with significant (P < 0.05) reduction in dopamine content in the corpus striatum. The immunohistochemical analysis revealed a marked decrease in tyrosine hydroxylase (TH) immunoreactivity in striatal neurons indicating the significant loss of dopaminergic neurons in substantia nigra (SN) following rotenone injection. However, chronic treatment with CVS restored the nerve terminals in the striatum from rotenone damage. CVS treatment improved the dopaminergic system function by restoring dopamine content in the striatum. CVS also improved the motor deformities clearly suggesting the neuroprotective function. Conclusion The results of the present study suggested CVS to be a safe and simple neuroprotective measure against neurodegenerative changes in PD and a promising noninvasive technique to overcome the motor symptoms associated with it. The findings could be useful for further investigations and clinical applications of CVS in the treatment of PD.
Collapse
Affiliation(s)
- J Thanalakshmi
- 1Department of Physiology, Saveetha Medical College Hospital, Saveetha Nagar, Thandalam,Chennai 602105, Tamil Nadu, India
| | - R Archana
- 1Department of Physiology, Saveetha Medical College Hospital, Saveetha Nagar, Thandalam,Chennai 602105, Tamil Nadu, India
| | - S Senthilkumar
- 2Department of Research and Development, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| | - R Shakila
- 3Department of Chemistry, Siddha Central Research Institute, Arumbakkam,Chennai, India
| | - N Pazhanivel
- 4Department of Veterinary Pathology, Madras Veterinary College, Chennai 602105, India
| | - S Subhashini
- 3Department of Chemistry, Siddha Central Research Institute, Arumbakkam,Chennai, India
| |
Collapse
|